Skip to main content

Full text of "USPTO Patents Application 10551504"

See other formats


(19) 


AUSTRALIAN PATENT OFFICE 


(11) Application No. fr\J 2004297111 A1 


(54) 


Title 

Anti-Mpl antibody 




/^h1 ^ B 
1° >) 


International Patent Classification(s) 

C07K. 16/28 (2006.01)20060101AFI2006040 

C07K 16/28 8BMEP 

PCT/JP2004/018506 




(21) 


Application No: 20042971 1 1 


(22) Application Date: 2004.12.10 


(87) 


WIPO No: WO05/056604 




(30) 
(31 ) 


Priority Data 

Number (321 Date 

2004-248323 2004.00.27 

2003- 415746 2003.12.12 

2004- 071763 2004.03.12 


(33) Country 
JP 
JP 
JP 


(43) 


Publication Date : 2005.06.23 




(71) 


Applicant(s) 

Chugai Seiyaku Kabushiki Kaisha 




(72) 


Inventor(s) 

Tsuchiya. Masayukij Nakano. Kiyotaka; 


Tsunoda. Hiroyuki; Hirata. Yuichi; Orita, Tetsuro 


(74) 


Agent/Attorney 

Spruson & Ferguson. Level 35, St Martins Tower 31 Market Street Sydney. NSW. 2000 



-1- 



1 



AU900W7III 



VERIFICATION OF TRANSLATION 

I, Mikiko Oyanagi 

of the Shimizu Patent Office, 6th Floor., Kantetsu Tsukuba-Science-City Bldg. 1-1-1, 
Oroshi-machi, Tsuchiura, Ibaraki, JAPAN 300-0847 

declare as follows: 



1 . That I am well acquainted with both the English and Japanese languages, and 

2. That the attached document is a true and correct translation, made by me to the best of 
my knowledge and belief, of:- 

International Application No. PCT/JP2004/0 18506 
(Publication No. WO2005/056604) 

Entitled: " ANTI-MPL ANTIBODY " 

Filed on December 10, 2004 

Mo(y ' I , fc> 

(Date) 




(Signature of translator) 
Mikiko Oyanagi 




•179179* 



I1N11IU 

•G00002* 



1 



DESCRIPTION 
ANT I -M PL ANTIBODY 

Technical Field 

5 The present invention relates to anti-Mpl antibodies. 

Background Art 

Thrombopoietin (TPO) is a factor that enhances the 
differentiation and maturation of megakaryocytes (platelet precursor 
10 cells) from hemopoietic stem cells into platelets. TPO also 
functions as a cytokine with an important role in the regulation of 
platelet number. TPO is converted into its active form through the 
cleavage of a TPO precursor comprising 353 amino acids. 

Mpl is a TPO receptor, and human Mpl molecules are known to exist 
15 in two forms comprising 572 and 635 amino acids. The human Mpl gene 
sequence has already been analyzed (see Non-Patent Document 1 and 
GenBank accession No. NM_005373) . 

Most cytokine receptors dimerize upon ligand binding, and 
transduce signals into cells. • It has been reported that TPO similarly 
20 binds to its own specific receptor MPL, which leads to dimerization 
of the receptor, thereby transducing signals into cells and exerting 
physiological effects (see Non-Patent Document 2) . 

Antibodies exhibiting agonistic activity have been reported 
among those antibodies that bind to receptors having the above 
25 features. 

For example, an antibody against the erythropoietin (EPO) 
receptor has been reported to substitute for erythropoietin function. 
The monovalent form (Fab) of the antibody is capable of binding to 
the EPO receptor but is unable to transduce signals. Thus, 
30 dimerization of the erythropoietin receptor via bivalent binding is 
assumed to be essential for signal transduction (see Non-Patent 
Document 3) . 

Antibodies that bind to Mpl and exhibit TPO agonistic activity 
have also been reported (see Non-Patent Documents 4 and 5) . This 
35 suggests that receptor dimerization is induced upon binding of a 
bivalent antibody with regards to MPL as well. 



2 



Meanwhile, a single-chain antibody (scFv) has been reported to 
exhibit TPO agonistic activity (see Patent Document 1) . However, it 
has been revealed that, the underlying mechanism of scFv exhibiting 
TPO agonistic activity is that a part of scFv diraerizes (diabody) 
5 and this diabody becomes the actual active unit (see Patent Documents 
2 to 4) . 

[Patent Document 1] US Patent No. 6342220 

[Patent Document 2] WO 01/79494 

[Patent Document 3] WO 02/33072 
10 [Patent Document 4] WO 02/33073 

[Non-Patent Document 1] Palacios et al., Cell, 1985, 41, 727-734 

[Non-Patent Document 2] Souyri et al., Cell, 1990, 63, 1137-1147 

[Non-Patent Document 3] Elliott, S. et al., J. Biol. Chem., 1996, 

271(40), 24691-24697 
15 [Non-Patent Document 4] Abe et al., Immunol. Lett., 1998, 61, 73-78 

[Non-Patent Document 5] Bi j ia Deng et al . , Blood, 1998, 92, 1981-1988 

Disclosure of the Invention 
Problems to Be Solved by the Invention 
20 The present invention was achieved in view of the above 

circumstances. An objective of the present invention is to provide 
novel anti-Mpl antibodies having TPO agonistic activity. 

Means to Solve the Problems 

25 The present inventors performed exhaustive research to solve 

the above objective. The present inventors prepared and purified 
anti-human Mpl antibody VB22B, and established a single-chain 
antibody expression system using genetic engineering techniques. 
Specifically, the variable region of anti-human Mpl antibody was first 

30 cloned, and a diabody expression vector pCXND3-VB22B db for the 
anti-human Mpl antibody was prepared. This pCXND3-VB22B db vector 
was then used to generate an expression vector pCXND3-VB22B sc(Fv)2 
for anti-human Mpl antibody sc(Fv)2. Anti-human Mpl sc(Fv)2 was 
expressed in CHO-DG44 cells using the expression vector pCXND3-VB22B 

35 sc(Fv)2, and then purified from the culture supernatant. In control 
experiments, VB22B diabody was transiently expressed in COS7 cells 



3 



using the above pCXND3-VB22B db vector, and then purified from the 
culture supernatant. 

In addition, VB22B diabody and VB22B sc (Fv) 2 were evaluated for 
their TPO-like agonistic activities. The results showed that VB22B 
5 diabody and VB22B sc(Fv)2 exhibit higher agonistic activities 
compared to VB22B IgG, and thus activities equivalent to or higher 
than that of the natural ligand, human TPO. 

Furthermore, the present inventors succeeded in preparing five 
types of humanized VB22B sc(Fv)2. The TPO-like agonistic activity 
10 was also proven to be unaltered by humanization. 

More specifically, the present invention provides the following 
(1) to (38): 

(1) an antibody comprising a single-chain polypeptide having 
binding activity against TPO receptor (Mpl), wherein said antibody 

15 comprises two heavy chain variable regions and two light chain 
variable regions; 

(2) the antibody of (1), wherein the two heavy chain variable 
regions and the two light chain variable regions are arranged in the 
order of heavy chain variable region, light chain variable region, 

20 heavy chain variable region, and light chain variable region from 
the N terminus of the single-chain polypeptide; 

(3) the antibody of (1) or (2), wherein the two heavy chain 
variable regions and the two light chain variable regions are linked 
by linkers; 

25 (4) the antibody of (3), wherein the linkers comprise 15 amino 

acids; 

(5) a chimeric antibody that binds to Mpl; 

(6) the antibody of (5), which is a humanized antibody; 

(7) the antibody of (5) or (6), which is a minibody; 
30 (8) an antibody that binds to soluble Mpl; 

(9) an antibody that binds to human Mpl and monkey Mpl; 

(10) an antibody having agonistic activity against human Mpl 
and monkey Mpl; 

(11) an antibody whose binding activity to soluble Mpl is KD 
35 = 10" 6 M or lower; 

(12) an antibody whose binding activity to soluble Mpl is KD 



4 



= 10 7 M or lower; 

(13) an antibody whose TPO agonistic activity is EC50 =100 nM 
or lower; 

(14) an antibody whose TPO agonistic activity is EC50 = 30 nM 
5 or lower; 

(15) an antibody whose TPO agonistic activity is EC50 = 10 nM 
or lower; 

(16) an antibody which comprises a heavy chain variable region, 
wherein said heavy chain variable regions comprises CDR1, CDR2 and 

10 CDR3 consisting of an amino acid sequence of any one of: 

[1) SEQ ID NOs: 3, 4, and 5 

[2] SEQ ID NOs: 6, 7, and 8 

[3] SEQ ID NOs: 9, 10, and 11 

14] SEQ ID NOs: 15, 16, and 17 
15 [5] SEQ ID NOs: 18, 19, and 20 

[6] SEQ ID NOs: 21, 22, and 23 

[7] SEQ ID NOs: 24, 25, and 26 

[8] SEQ ID NOs: 27, 28, and 29 

[9] SEQ ID NOs: 30, 31, and 32 



20 


[10] 


SEQ 


ID 


NOs: 


33, 


34, 


and 


35 




[11] 


SEQ 


ID 


NOs: 


36, 


37, 


and 


38 




[12] 


SEQ 


ID 


NOs : 


39, 


40, 


and 


41 




[13] 


SEQ 


ID 


NOs: 


42, 


43, 


and 


44 




[14] 


SEQ 


ID 


NOs : 


48, 


49, 


and 


50 


25 


[15] 


SEQ 


ID 


NOs : 


51, 


52, 


and 


53 




[16] 


SEQ 


ID 


NOs : 


54, 


55, 


and 


56 




[17] 


SEQ 


ID 


NOs: 


57, 


58, 


and 


59; 



(17) an antibody which comprises a light chain variable region, 
wherein said light chain variable region comprises CDRl, CDR2 and 
30 CDR3 consisting of an amino acid sequence of any one of: 

[1] SEQ ID NOs: 60, 61, and 62 

[2] SEQ ID NOs: 63, 64, and 65 

[3] SEQ ID NOs: 78, 79, and 80 

[4] SEQ ID NOs: 84, 85, and 86 
35 [5] SEQ ID NOs: 93, 94, and 95 

[6] SEQ ID NOs: 96, 97, and 98 



[7] SEQ ID NOs: 102, 103, and 104 
[8] SEQ ID NOs : 108, 109, and 110 
[9] SEQ ID NOs: 111, 112, and 113 
[10] SEQ ID NOs: 114, 115, and 116; 

(IB) an antibody that comprises a heavy chain variable region 
and a light chain variable region of any one of: 

[1] a heavy chain variable region that comprises CDR1, CDR2 , 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 3, 4, and 5, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 60, 61, and 62; 

[2] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 6, 7, and 8, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[3] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 9, 10, and 11, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[4] a heavy chain variable region that comprises CDRl, CDR2 , 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 15, 16, and 17, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[5] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 18, 19, and 20, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[6] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 21, 22, and 23, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 78, 79, and 80; 



6 



[7] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs : 24, 25, and 26, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
5 of SEQ ID NOs: 63, 64, and 65; 

[8] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 27, 28, and 29, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
10 of SEQ ID NOs: 84, 85, and 86; 

[9] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 30, 31, and 32, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequence consisting 
15 of SEQ ID NOs: 63, 64, and 65; 

[10] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 33, 34, and 35, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
20 of SEQ ID NOs: 63, 64, and 65; 

[11] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 36, 37, and 38, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
25 of SEQ ID NOs: 93, 94, and 95; 

[12] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 39, 40, and 41, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
30 of SEQ ID NOs: 96, 97, and 98; 

[13] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 42, 43, and 44, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
35 of SEQ ID NOs: 78, 79, and 80; 

[14] a heavy chain variable region that comprises CDR1, CDR2, 



7 



and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 45, 46, and 47, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 102, 103, and 104; 
5 [15] a heavy chain variable region that comprises CDR1, CDR2, 

and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 48, 49, and 50, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

10 [16] a heavy chain variable region that comprises CDR1, CDR2, 

and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 51, 52, and 53, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 108, 109, and 110, 

15 [17] a heavy chain variable region that comprises CDR1, CDR2, 

and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 54, 55, and 56, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 111, 112, and 113; 

20 [18] a heavy chain variable region that comprises CDR1, CDR2, 

and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 57, 58, and 59, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 each comprising the amino acid sequences 
consisting of SEQ ID NOs: 114, 115, and 116; 

25 (19) an antibody that comprises a heavy chain variable region 

comprising the amino acid sequence of SEQ ID NO: 118; 

(20) an antibody that comprises a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 120; 

(21) an antibody that comprises a heavy chain variable region 
30 comprising the amino acid sequence of SEQ ID NO: 118 and a light chain 

variable region comprising the amino acid sequence of SEQ ID NO: 120; 

(22) an antibody comprising the amino acid sequence of SEQ ID 
NO: 122 or 264; 

(23) an antibody that comprises a heavy chain variable region, 
35 wherein said heavy chain variable region comprises FR1, FR2, FR3, 

and FR4 consisting of amino acid sequences of any one of: 



8 



[1] 


SEQ 


ID NOs: 230, 232, 234, 


and 236 


[2] 


SEQ 


ID NOs: 265, 267, 269, 


and 271 


[3] 


SEQ 


ID NOs: 279, 281, 283, 


and 285 


[4] 


SEQ 


ID NOs: 298, 299, 300, 


and 301 


[5] 


SEQ 


ID NOs: 298, 299, 306, 


and 301. 


(24) 


an 


antibody comprising a 


light chain variable region, 



wherein said light chain variable region comprises FR1, FR2, FR3, 
and FR4 consisting of amino acid sequences of any one of: 

[1] SEQ ID NOs: 239, 241, 243, and 245 
10 [2] SEQ ID NOs: 272, 274, 276, and 278 

[3] SEQ ID NOs: 302, 303, 304, and 305 

[4] SEQ ID NOs: 302, 307, 308, and 305; 

(25) an antibody that comprises a heavy chain variable region 
and a light chain variable region of any one of: 

15 [1] a heavy chain variable region which comprises FR1, FR2, FR3, 

and FR4 having the amino acid sequences consisting of SEQ ID NOs: 
230, 232, 234, and 236, and a light chain variable region which 
comprises FR1, FR2, FR3, and FR4 having the amino acid sequences 
consisting of SEQ ID NOs: 239, 241, 243, and 245; 

20 [2] a heavy chain variable region which comprises FR1 , FR2, FR3, 

and FR4 having the amino acid sequences consisting of SEQ ID NOs: 
265, 267, 269, and 271, and a light chain variable region which 
comprises FR1, FR2, FR3, and FR4 having the amino acid sequences 
consisting of SEQ ID NOs: 272, 274, 276, and 278; 

25 [3] a heavy chain variable region which comprises FR1 , FR2 , FR3, 

and FR4 having the amino acid sequences consisting of SEQ ID NOs: 
279, 281, 283, and 285, and a light chain variable region which 
comprises FR1, FR2, FR3, and FR4 having the amino acid sequences 
consisting of SEQ ID NOs: 272, 274, 276, and 278; 

30 [4] a heavy chain variable region which comprises FR1, FR2, FR3, 

and FR4 having the amino acid sequences consisting of SEQ ID NOs: 
298, 299, 300, and 301, and a light chain variable region which 
comprises FR1, FR2, FR3, and FR4 having the amino acid sequences 
consisting of SEQ ID NOs: 302, 303, 304, and 305; 

35 [5] a heavy chain variable region which comprises FR1, FR2, FR3, 

and FR4 having the amino acid sequences consisting of SEQ ID NOs: 



9 



298, 299, 306, and 301,' and a light chain variable region which 
comprises FR1, FR2 , FR3, and FR4 having the amino acid sequences 
consisting of SEQ ID NOs: 302, 307, 308, and 305; 

(26) an antibody that comprises a heavy chain variable region, 
5 wherein said heavy chain variable region comprises the amino acid 

sequence of SEQ ID NO: 229, 256, 262, 289, or 295; 

(27) an antibody that comprises a light chain variable region, 
wherein said light chain variable region comprises the amino acid 
sequence of SEQ ID NO: 238, 258, 291, or 297; 

10 (28) -an antibody that comprises a heavy chain variable region 

and a light chain variable region of any one of: 

[1] a heavy chain variable region comprising the amino acid 
sequence of SEQ ID NO: 229, and a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 238; 
15 [2] a heavy chain variable region comprising the amino acid 

sequence of SEQ ID NO: 256, and a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 258; 

[3] a heavy chain variable region comprising the amino acid 
sequence of SEQ ID NO: 262, and a light chain variable region 
20 comprising the amino acid sequence of SEQ ID NO: 258; 

[4] a heavy chain variable region comprising the amino acid 
sequence of SEQ ID NO: 289, and a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 291; 

[5] a heavy chain variable region comprising the amino acid 
25 sequence of SEQ ID NO: 295, and a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 297; 

(29) an antibody that comprises the amino acid sequence of SEQ 
ID NO: 2, 254, 260, 287, or 293; 

(30) an antibody having an activity equivalent to that of an 
30 antibody of any one of (16) to (29), wherein said antibody comprises 

the amino acid sequence set forth in any one of (16) to (29) , in which 
one or more amino acids have been substituted, deleted, added and/or 
inserted; 

(31) an antibody that recognizes an epitope recognized by an 
35 antibody of any one of (16) to (30); 

(32) an antibody that recognizes the region of amino acids. 26 



10 



to 27 4 of human Mpl; 

(33) an antibody of any one of (1) to (32), which has TPO 
agonistic activity; 

(34) a polynucleotide encoding an antibody of any one of (1) 

to (33) ; 

(35) a polynucleotide hybridizing to the polynucleotide of (34) 
under stringent conditions, wherein said polynucleotide encodes an 
antibody having activity equivalent to that of an antibody of any 
one of (1) to (33) ; 

(36) a vector comprising the polynucleotide of (34) or (35); 

(37) a host cell that carries the polynucleotide of (34) or (35) , 
or the vector of (36) ; and 

(38) a pharmaceutical composition comprising an antibody of any 
one of (1) to (33') . 

Brief Description of the Drawings 

Fig. 1 demonstrates the strategy for preparing single-chain 
antibody sc(Fv)2. 

Fig. 2 illustrates the assessment of VB22B sc(Fv)2 binding 
activity using an Mpl-expressing CHO cell line. Purified VB22B 
sc(Fv)2 was used. 

Fig. 3 illustrates the assessment of VB22B antibody agonistic 
activity using BaF3-human Mpl. 

Fig. 4 illustrates the assessment of VB22B antibody agonistic 
activity using BaF3-monkey Mpl. 

Fig. 5 illustrates the assessment of VB22B antibody agonistic 
activity using M-07e. 

Fig. 6 shows the amino acid sequences of anti-human Mpl 
antibodies (H chains) that exhibit higher agonistic activities when 
converted into minibodies. 

Fig. 7 shows the amino acid sequences of anti-human Mpl 
antibodies (L chains) which exhibit higher agonistic activities when 
converted into minibodies. 

Fig. 8 illustrates the binding activity assessment of AB317 
diabody using Mpl-expressing CHO cells. Both VB22B diabody (solid 
line) and AB317 diabody (broken line) were obtained from C0S7 culture 



11 



supernatants . 

Fig. 9 illustrates the agnostic activity assessment of AB324 
and AB317 diabodies using BaF3-human Mpl . 

Fig. 10 illustrates the agnostic activity assessment of AB324 
and AB317 diabodies using BaF3-monkey Mpl. 

Fig. 11 illustrates the agnostic activity assessment of AB324 
and AB317 diabodies using BaF3-mouse Mpl. 

Fig. 12 shows the agonistic activities of diabodies in 
BaF3-human Mpl cells. The X-axis shows OD at 450/655 rati, and the 
Y-axis represents concentration. 

Fig. 13 shows the agonistic activities of diabodies in 
BaF3-human Mpl (G305C) cells. The X-axis shows OD at 450/655 nm, and 
the Y-axis represents concentration. 

Fig. 14 shows the agonistic activities of TA136 db and TA136 
sc(Fv)2 in 8aF3-human Mpl cells. The X-axis shows OD at 450/655 nm 
and the Y-axis represents concentration. 

Fig. 15 shows the agonistic activities of TA136 db and TA136 
sc(Fv)2 in BaF3-human Mpl (G305C) cells. The X-axis shows OD at 
450/655 nm, and the Y-axis represents concentration. 

Fig. 16 shows the agonistic activities of TA136 db and TA136 
sc{Fv)2 in BaF3-human Mpl (C769T) cells. The X-axis shows OD at 
450/655 nm, and the Y-axis represents concentration. 

Fig. 17 shows the agonistic activities of TA136 db and TA136 
sc(Fv)2 in BaF3-human Mpl (C823A) cells. The X-axis shows OD at 
450/655 nm, and the Y-axis represents concentration. 

Fig. 18 shows the positions of FRs and CDRs in humanized heavy 
chain seguences (hVB22B p-z, hVB22B g-e, hVB22B e, hVB22B u2-wz4, 
and hVB22B q-wz5:VH), and humanized light chain sequences (hVB22B 
p-z, hVB22B g-e, hVB22B e, hVB22B u2-wz4, and hVB22B q-wz5:VL). 

Fig. 19 shows the TPO-like agonistic activities of murine VB22B 
sc(Fv)2, hVB22B e sc(Fv)2, and hVB22B g-e sc(Fv)2 in BaF3-human Mpl. 
The X-axis shows absorbance ratio (450 nm/655 nm) , and the Y-axis 
represents concentration. 

Fig. 20 shows the TPO-like agonistic activities of murine VB22B 
sc(Fv)2, hVB22B p-z sc (Fv) 2, and hVB22B u2-wz4 sc(Fv)2in BaF3-human 
Mpl. The X-axis shows absorbance ratio {450 nm/655 nm) , and the Y-axis 



12 



represents concentration. 

Fig. 21 shows the TPO-like agonistic activities of murine VB22B 
sc(Fv}2 and hVB22B q-wz5 sc(Fv)2 in BaF3-human Mpl . The X-axis shows 
absorbance ratio (450 nm/655 nm) , and the Y-axis represents 
5 concentration. 

Best Mode for Carrying Out the Invention 

The present invention provides antibodies that bind to the TPO 
receptor (Mpl) . 

10 The antibodies of the present invention comprise all types of 

antibodies, including antibodies with modified amino acid sequences, 
such as minibodies, humanized antibodies, and chimeric antibodies; 
antibodies that have been modified by binding with other molecules 
(for example, polymers such as polyethylene glycol) ; and antibodies 

15 whose sugar chains have been modified. 

Mpl of the present invention may be a mutant receptor. A mutant 
receptor of the present invention is usually a receptor that exists 
at a frequency lower than 50%, preferably lower than 20%, more 
preferably lower than 10%, and even more preferably lower than 1%. 

20 The frequency is generally calculated using randomly selected 
subjects. However, the frequency may vary depending on the country, 
area, sex, and such. Therefore, the frequency may also be calculated, 
for example, within a defined country or area, such as Japan, the 
United States, and Europe, or calculated for one sex. When there are 

25 two or more mutations in a receptor, the frequency may be calculated 
for multiple mutation sites or for any one of the mutation sites. 
Mutant receptors are preferably evaluated by a frequency as described 
above. However, mutant receptors can also be evaluated, for example, 
by their signal transducing ability and such. Specifically, for 

30 example, when two different receptors are present, the one with 
stronger transducing signals upon natural ligand-binding maybe be 
used as a non-mutant type receptor, and the one with weaker transducing 
signals as a mutant receptor. 

In one embodiment, the mutant receptors of the present invention 

35 comprise receptors that are associated with disease onset. The 
phrase "mutant receptors associated with disease onset" means that 



13 



the loss of reactivity to a natural ligand becomes part of the reason 
that triggers disease onset. In the present invention, the mutant 
receptor may be a contributing factor, but not necessarily the sole 
factor triggering disease onset. Many reports have been previously 
5 published that describe the association of mutant receptors with 
disease onset. In addition to those that have been reported, 
associations of mutant receptors and disease onset can also be 
identified by statistical analysis methods (for example, correlation 
analyses) . Correlation analyses, also called "case control studies", 

10 are well known to those skilled in the art (for example, Nishimura, 
Y . , 1991, "Statistical analysis of polymorphisms", Saishin Igaku, 
46:909-923; Oka, A. et al . , 1990, Hum. Mol. Genetics 8, 2165-2170; 
Ota, M. et al., 1999, Am. J. Hum. Genet. 64, 1406-1410; Ozawa, A. 
et al., 1999, Tissue Antigens 53, 263-268). For example, the 

15 correlation between a mutant receptor and a disease can be studied 
by computing the freguency of the mutant receptor in patients and 
healthy subjects, and testing whether the patient population has a 
higher mutant receptor freguency. Typically, differences in 
frequency are evaluated using the fc-test. X is obtained by the 

20 equation % 2 - 2 (observed value - expected value ) 2 /expected value. A 
p value is obtained from the X 2 value determined. Based on this p 
value, it can be determined whether there is a correlation between 
the mutant receptor and the disease. For example, when p<0.05, the 
mutant receptor is considered to correlate with the disease. Mutant 

25 thrombopoietin (TPO) receptors have already been reported (Matthias 
Ballmaier et al., 2001, BLOOD, 97 (1), 139; and others). 

It is preferable that the antibodies of the present invention 
have agonistic activity against Mpl . 

In a preferred embodiment, the antibodies of the present 

30 invention comprise, for example, minibodies. 

The minibodies comprise antibody fragments lacking portions of 
the whole antibody (for example, whole IgG) . The minibodies are not 
particularly limited as long as they have binding activity to their 
antigens. The minibodies of the present invention have' higher 

35 activities compared to their corresponding whole antibodies. There 
are no particular limitations on the antibody fragments of the present 



14 



invention as long as they are portions of the whole antibody, and 
preferably contain heavy chain variable regions (VH) and/or light 
chain variable regions (VL) . The amino acid sequences of VH or VL 
may contain substitutions, deletions, additions and/or insertions. 
5 Furthermore, the antibody fragment may also lack portions of VH or/and 
VL, as long as it has binding ability to its antigen. In addition, 
the variable regions may be chimerized or humanized. Such antibody 
fragments include, for example, Fab, Fab', F(ab')z, and Fv. An 
example of a minibody includes Fab, Fab', F(ab') 2 , Fv, scFv 

10 (single-chain Fv) , diabody, and sc(Fv)2 (single-chain (Fv)2). 

Herein, an "Fv" fragment is the smallest antibody fragment and 
contains a complete antigen recognition site and a binding site. The 
"Fv" fragment is a dimer (VH-VL dimer) in which a single VH and a 
single VL are strongly linked by a non-covalent bond. The three 

15 complementarity-determining regions (CDRs) of each of the variable 
regions interact with each other to form an antigen-binding site on 
the surface of the VH-VL dimer. Six CDRs confer the antigen-binding 
site of an antibody. However, a single variable region (or a half 
of Fv containing only three CDRs specific to an antigen) alone is 

20 also capable of recognizing and binding an antigen although its 
affinity is lower than the affinity of the entire binding site. 

scFv contains the VH and VL regions of an antibody, and these 
regions exist on a single polypeptide chain. Generally, an Fv 
polypeptide further contains a polypeptide linker between VH and VL, 

25 and therefore an scFv can form a structure required for antigen binding . 
See, Pluckthun "The Pharmacology of Monoclonal Antibodies" Vol. 113 
(Rosenburg and Moore eds. (Springer Verlag, New York, pp. 269-315, 
1994) for the review of scFv. In the present invention, linkers are 
not especially limited as long as they do not inhibit expression of 

30 antibody variable regions linked at both ends of the linkers. 

The term "diabody" refers to a bivalent antibody fragment 
constructed by gene fusion (Holliger P et al . , 1993, Proc. Natl . Acad. 
Sci. USA 90: 6444-6448; EP 404,097; WO 93/11161 and others). 
Diabodies are dimers comprising two polypeptide chains, where each 

35 polypeptide chain comprises a VL and a VH connected with a linker 
short enough to prevent interaction of these two domains, for example, 



15 



a linker of about five residues. The VL and VH encoded on the same 
polypeptide chain will form a dimer because the linker between them 
is too short to form a single-chain variable region fragment. As a 
result, the polypeptide chains form a dimer, and thus the diabody 
5 has two antigen binding sites. 

sc (Fv) 2 is a single-chain minibody produced by linking two units 
of VH and two units of VL with linkers and such (Hudson et ai., 1999, 
J Immunol. Methods 231:177-189). sc(Fv)2 exhibits a particularly 
high agonistic activity compared to the whole antibody and other 
10 minibodies. sc (Fv) 2 can be produced, for example, by linking two scFv 
molecules . 

In a preferable antibody, the two VH units and two VL units are 
arranged in the order of VH, VL, VH, and VL 
( [VH] -linker- [VL] -linker- [VH] -linker- [VL] ) beginning from the N 

15 terminus of a single-chain polypeptide. 

The order of the two VH units and two VL units is not limited 
to the above arrangement, and they may be arranged in any order. 
Examples of the arrangements are listed below. 
[VL] -linker- [VH] -linker- [VH] -linker- [VL] 

2 0 [VH] -linker- [VL] -linker- [VL] -linker- [VH] 
[VH] -linker- [VH] -linker- [VL] -linker- [VL] 
[VL] -linker- [VL] -linker- [VH] -linker- [VH] 
[VL] -linker- [VH] -linker- [VL] -linker- [VH] 

The linkers to be used for linking the variable regions of an 

25 antibody comprise arbitrary peptide linkers that can be introduced 
by genetic engineering, synthetic linkers, and linkers disclosed in, 
for example, Holliger, P. et al . , Protein Engineering, 9(3), 299-305, 
1996. Peptide linkers are preferred in the present invention. There 
are no limitations as to the length of the peptide linkers. The length 

30 can be selected accordingly by those skilled in the art depending 
on the purpose, and is typically 1-100 amino acids, preferably 3-50 
amino acids, more preferably 5-30 amino acids, and even more 
preferably 12-18 amino acids (for example, 15 amino acids) . 
For example, such peptide linkers include: 

35 Ser 

Gly • Ser 



16 



GlyGly Ser 
Ser-Gly ■ Gly 
Gly Gly Gly Ser 
Ser- Gly Gly Gly 
5 Gly Gly Gly Gly Ser 
Ser • Gly Gly • Gly Gly 
Gly Gly Gly Gly Gly Ser 
Ser • Gly Gly Gly Gly Gly 
Gly Gly Gly • Gly Gly Gly Ser 
10 Ser-Gly Gly GlyGly Gly Gly 

(GlyGly Gly Gly Ser) „ 

(Ser-Gly-Gly Gly-Gly) n 

where n is. an integer of 1 or larger. The lengths and sequences of 
peptide linkers can be selected accordingly by those skilled in the 

15 art depending on the purpose. 

In an embodiment of the present invention, a particularly 
preferable sc(Fv)2 includes, for example, the sc(Fv)2 below. 
[VH] -peptide linker (15 amino acids) - [VL] -peptide linker (15 amino 
acids) - [VH] -peptide linker (15 amino acids ) - [VL] 

20 Synthetic linkers (chemical crosslinking agents) include 

crosslinking agents routinely used to crosslink peptides, for example, 
N-hydroxy succinimide (NHS), disuccinimidyl suberate (DSS), 
bis ( succinimidyl ) suberate (BS 3 ) , dithiobis ( succinimidyl 
propionate) (DSP), dithiobi s ( succinimidyl propionate) (DTSSP) , 

25 ethylene glycol bis ( succinimidyl succinate) (EGS) , ethylene glycol 
bis ( sulf osuccinimidyl succinate) (sulfo-EGS), disuccinimidyl 
tartrate (DST), disulf osuccinimidyl tartrate (sulfo-DST) , 
bis [ 2- ( succinimidoxycarbonyloxy) ethyl ] sulf one (BSOCOES) , and 
bis [ 2- ( succinimidoxycarbonyloxy) ethyl ] sulfone (sulf o-BSOCOES) . 

30 These crosslinking agents are commercially available. 

In general, three linkers are required to link four antibody 
variable regions together. The linkers to be used may be of the same 
type or different types. In the present invention, a preferable 
minibody is a diabody, even more preferably, an sc(Fv)2. Such a 

35 minibody can be prepared by treating an antibody with an enzyme, for 
example, papain or pepsin, to generate antibody fragments, or by 



17 



constructing DNAs encoding those antibody fragments and introducing 
there into expression vectors, followed by expression in an appropriate 
host cell (see, for example, Co, M. S. et al., 1994, J. Immunol. 152, 
2968-2976; Better, M. and Horwitz, A. H., 1989, Methods Enzymol. 178, 
476-496; Pluckthun, A. andSkerra, A., 1989, Methods Enzymol. 178, 
497-515; Lamoyi, E . , 1986, Methods Enzymol. 121, 652-663; Rousseaux, 
J. et al. , 1986, Methods Enzymol . 121, 663-669; Bird, R. E. and Walker, 
B. W., 1991, Trends Biotechnol. 9, 132-137). 

An antibody having exceedingly high agonistic activity can be 
prepared by reducing the molecular weight of a full-length antibody, 
particularly by converting it into an sc(Fv)2. 

In a preferred embodiment, the antibodies of the present 
invention comprise modified antibodies, such as chimeric antibodies 
and humanized antibodies that bind to Mpl . These modified antibodies 
can be produced by known methods. 

Chimeric antibodies are antibodies prepared by combining 
sequences derived from different animal species, and include for 
example, antibodies comprising the heavy chain and light chain 
variable regions of a murine antibody, and the heavy chain and light 
chain constant regions of a human antibody. Chimeric antibodies can 
be prepared by known methods . For example, a DNA encoding the V region 
of an antibody is linked to a DNA encoding the C region of a human 
antibody, and the construct is inserted into an expression vector 
and introduced into a host to produce chimeric antibodies. 

Humanized antibodies are also referred to as "reshaped human 
antibodies". Such a humanized antibody is obtained by transferring 
the complementarity-determining region (CDR) of an antibody derived 
from a non-human mammal, for example mouse, to the 
complementarity-determining region of a human antibody, and the 
general gene recombination procedure for this is also known (see 
European Patent Application No. 125023 and WO 96/02576) . 

Specifically, a DNA sequence designed to link a murine antibody 
CDR to the framework region (FR) of a human antibody can be synthesized 
by PCR, using primers prepared from several oligonucleotides 
containing overlapping portions of both CDR and FR terminal regions 
(see methods described in WO 98/13388) . 



18 



The human antibody framework region to be linked by CDR is 
selected in order to form a favorable antigen-binding site in the 
complementarity-determining region. Amino acids of the framework 
region in the antibody variable region may be substituted, as 
5 necessary, for the complementarity-determining region of the reshaped 
human antibody to form a suitable antigen-binding site (Sato, K. et 
al., 1993, Cancer Res. 53, 851-856). 

The constant region of a human antibody is used as the constant 
region of a chimeric antibody or humanized antibody. For example, 
10 Cyl, Cy2, Cy3, and Cy4 can be used as the H chain, and Ck and CX. can 
be used as the L chain. The human antibody constant region may be 
modified to improve the antibody or the stability of the antibody 
production . 

Generally, chimeric antibodies comprise. the variable region of 
15 an antibody from a non-human mammal and the constant region derived 
from a human antibody. On the other hand, humanized antibodies 
comprise the complementarity-determining region of an antibody from 
a non-human mammal, and the framework region and constant region 
derived from a human antibody. 
20 In addition, after a chimeric antibody or a humanized antibody 

is prepared,' amino acids in the variable region (for example, FR) 
and the constant region may be replaced with other amino acids, and 
such . 

The origin of the variable regions in chimeric antibodies or 
25 that of the CDRs in humanized antibodies is not particularly limited, 
and may be derived from any type of animal. For example, sequences 
of murine antibodies, rat antibodies, rabbit antibodies, camel 
antibodies may be used. 

In general, it is difficult to chimerize or humanize an antibody 
30 without losing the agonistic activity of the original antibody. 
Nevertheless, the present invention succeeded in preparing humanized 
antibodies having agonistic activity equivalent to that of the 
original murine antibody. 

A preferred humanized antibody of the present invention is an 
35 antibody comprising a heavy chain variable region that comprises the 
amino acid sequence of SEQ ID NO: 229 (humanized heavy chain sequence : 



19 



hVB22B p-z VH) , SEQ ID NO: 256 (humanized heavy chain sequence: hVB22B 
g-e VH) , SEQ ID NO: 262 (humanized heavy chain sequence: hVB22B e 
VH) , SEQ ID NO: 289 (humanized heavy chain sequence: hVB22B u2-wz4 
VH) , or SEQ ID NO: 295 (humanized heavy chain sequence: hVB22B q-wz5 
5 VH) ; or an antibody comprising a light chain variable region that 
comprises the amino acid sequence of SEQ ID NO: 238 (humanized light 
chain hVB22B p-z VL) , SEQ ID NO: 258 (humanized light chain hVB22B 
g-e VL or hVB22B e VL) , SEQ ID NO: 291 (humanized light chain hVB22B 
u2-wz4 VL) , or SEQ ID NO: 297 (humanized light chain hVB22B q-wz5 
10 VL) . In particular, a preferred antibody is an antibody comprising 
a heavy chain variable region and a light chain variable region of 
any one of (1) to (5) indicated below: 

(1) a heavy chain variable region comprising the amino acid sequence 
of SEQ ID NO: 229, and a light chain variable region comprising the 

15 amino acid sequence of SEQ ID NO: 238; 

(2) a heavy chain variable region comprising the amino acid sequence 
of SEQ ID NO: 256, and a light chain variable region comprising the 
amino acid sequence of SEQ ID NO: 258; 

(3) a heavy chain variable region comprising the amino acid sequence 
20 of SEQ ID NO: 262, and a light chain variable region comprising the 

amino acid sequence of SEQ ID NO: 258; 

(4) a heavy chain variable region comprising the amino acid sequence 
of SEQ ID NO: 289, and a light chain variable region comprising the 
amino acid sequence of SEQ ID NO: 291; and 

25 (5) a heavy chain variable region comprising the amino acid sequence 

of SEQ ID NO: 295, and a light chain variable region comprising the 

amino acid sequence of SEQ ID NO: 297. 

Such antibodies include, for example, antibodies comprising the 

amino acid sequence of SEQ ID NO: 2, 254 , 260, 287, or 293 (humanized 
30 sc(Fv)2 sequence (hVB22B p-z sc(Fv)2, hVB22B g-e sc(Fv)2, hVB22B e 

sc(Fv)2, hVB22B u2-wz4, or hVB22B q-wz5) . 

The nucleotide sequence of hVB22B p-z VH is shown in SEQ ID NO: 

228; the nucleotide sequence of hVB22B g-e VH is shown in SEQ ID NO: 

255; the nucleotide sequence of hVB22B e VH is shown in SEQ ID NO: 
35 261; the nucleotide sequence of hVB22B u2-wz4 VH is shown in SEQ ID 

NO: 288; the nucleotide sequence of hVB22B q-wz5 VH is shown in SEQ 



20 



ID NO: 294; the nucleotide sequence of hVB22B p-z VL is shown in SEQ 
ID NO: 237; the nucleotide sequences of hVB22B g-e VL and hVB22B e 
VL are shown in SEQ ID NO: 257; the nucleotide sequence of hVB22B 
u2-wz4 VL is shown in SEQ ID NO: 290; and the nucleotide sequence 
of hVB22B q-wz5 VL is shown in SEQ ID NO: 296. 

In the amino acid sequence of SEQ ID NO: 229 (humanized heavy 
chain sequence: hVB22B p-z VH) , SEQ ID NO: 256 (humanized heavy chain 
sequence: hVB22B g-e VH) , SEQ ID NO: 262 (humanized heavy chain 
sequence: hVB22B e VH) , SEQ ID NO: 289 (humanized heavy chain sequence: 
hVB22B u2-wz4 VH) , or SEQ ID NO: 295 (humanized heavy chain sequence: 
hVB22B q-wz5 VH) , 

amino acids 31-35 correspond to CDR1; 
amino acids 50-66 correspond to CDR2; 
amino acids 99-107 correspond to CDR3; 
amino acids 1-30 correspond to FR1; 
amino acids 36-4 9 correspond to FR2; 
amino acids 67-98 correspond to FR3; and 
amino acids 108-118 correspond to FR4 . 

In the amino acid sequence of SEQ ID NO: 238 (humanized light 
chain sequence: hVB22B p-z VL) , SEQ ID NO: 258 (humanized light chain 

sequence: hVB22B g-e VL or hVB22B e VL) , SEQ ID NO: 291 (humanized 

light chain sequence: hVB22B u2-wz4 VL) , or SEQ ID NO: 297 (humanized 

light chain sequence: hVB22B q-wz5 VL) , 

amino acids 24-39 correspond to CDRl; 

amino acids 55-61 correspond to CDR2; 

amino acids 94-102 correspond to CDR3; 

amino acids 1-23 correspond to FR1; 

amino acids 40-54 correspond to FR2 ; 

amino acids 62-93 correspond to FR3; and 

amino acids 103-112 correspond to FR4 . 

In the present invention, SEQ ID NOs of the CDRs and FRs in the 

hVB22B p-z VH sequence are shown below: 

hVB22B p-z VH: FR1/SEQ ID NO: 230 

hVB22B p-z VH: CDR1/SEQ ID NO: 36 

hVB22B p-z VH: FR2/SEQ ID NO: 232 

hVB22B p-z VH: CDR2/SEQ ID NO: 37 



21 



hVB22B p-z VH: FR3/SEQ ID NO: 234 

hVB22B p-z VH: CDR3/SEQ ID NO: 38 

hVB22B p-z VH: FR4/SEQ ID NO: 236. 

In the present invention, SEQ ID NOs of the CDRs and FRs in the 
5 hVB22B p-z VL sequence are shown below: 

hVB22B p-z VL: FR1/SEQ ID NO: 239 

hVB22B p-Z VL: CDR1/SEQ ID NO: 93 

hVB22B p-z VL: FR2/SEQ ID NO: 241 

hVB22B p-z VL: CDR2/SEQ ID NO: 94 
10 hVB22B p-z VL: FR3/SEQ ID NO: 243 

hVB22B p-z VL: CDR3/SEQ ID NO: 95 

hVB22B p-z VL: FR4/SEQ ID NO: 245. 

In the present invention, SEQ ID NOs of the CDRs and FRs in the 

hVB22B g-e VH sequence are shown below: 
15 hVB22B g-e VH: FR1/SEQ ID NO: 265 

hVB22B g-e VH: CDR1/SEQ ID NO: 36 

hVB22B g-e VH: FR2/SEQ ID NO: 267 

hVB22B g-e VH: CDR2/SEQ ID NO: 37 

hVB22B g-e VH: FR3/SEQ ID NO: 269 
20 1YVB22B g-e VH: CDR3/SEQ ID NO: 38 

hVB22B g-e VH: FR4/SEQ ID NO: 271. 

In the present invention, SEQ ID NOs of the CDRs and FRs in the 

1YVB22B g-e VL sequence are shown below: 

hVB22B g-e VL: FR1/SEQ ID NO: 272 
25 hVB22B g-e VL: CDR1/SEQ ID NO: 93 

hVB22B g-e VL: FR2/SEQ ID NO: 274 

hVB22B g-e VL: CDR2/SEQ ID NO: 94 

hVB22B g-e VL: FR3/SEQ ID NO: 276 

hVB22B g-e VL: CDR3/SEQ ID NO: 95 
30 hVB22B g-e VL: FR4/SEQ ID NO: 278. 

In the present invention, SEQ ID NOs of the CDRs and FRs in the 

hVB22B e VH sequence are shown below: 

hVB22B e VH: FR1/SEQ ID NO: 279 

hVB22B e VH : CDRl/SEQ ID NO:' 36 
35 hVB22B e VH: FR2/SEQ ID NO: 281 

hVB22B e VH : CDR2/SEQ ID NO: 37 



22 



hVB22B e VH: FR3/SEQ ID NO: 283 

hVB22B e VH: CDR3/SEQ ID NO: 38 

hVB22B e VH: FR4/SEQ ID NO: 285. 

In the present invention, SEQ ID NOs of the CDRs and FRs in the 
5 hVB22B e VL sequence are shown below: 

hVB22B e VL: FR1/SEQ ID NO: 272 

hVB22B e VL : CDR1/SEQ ID NO: 93 

hVB22B e VL: FR2/SEQ ID NO: 274 

hVB22B e VL: CDR2/SEQ ID NO: 94 
10 hVB22B e VL: FR3/SEQ ID NO: 276 

hVB22B e VL: CDR3/SEQ ID NO: 95. 

hVB22B e VL: FR4/SEQ ID NO: 278. 

In the present invention, SEQ ID NOs of the CDRs and FRs in the 

hVB22B u2-wz4 VH sequence are shown below: 
15 hVB22B u2-wz4 VH : FR1/SEQ ID NO: 298 

hVB22B u2-wz4 VH: CDR1/SEQ ID NO: 36 

hVB22B u2-wz4 VH: FR2/SEQ ID NO: 299 

hVB22B u2-wz4 VH: CDR2/SEQ ID NO: 37 

hVB22B u2-wz4 VH: FR3/SEQ ID NO: 300 
20 hVB22B u2-wz4 VH: CDR3/SEQ ID NO: 38 

hVB22B u2-wz4 VH : FR4/SEQ ID NO: 301. 

In the present invention, SEQ ID NOs of the CDRs and FRs in the 





hVB22B 


u2 


-wz 4 


VL 


sequence are shown b 




hVB22B 


u2 


-wz4 


VL: 


FR1/SEQ ID NO: 


302 


25 


hVB22B 


u2 


-wz4 


VL: 


CDR1/SEQ ID NO: 


93 




hVB22B 


u2 


-wz4 


VL: 


FR2/SEQ ID NO: 


303 




hVB22B 


u2 


-wz4 


VL: 


CDR2/SEQ ID NO: 


94 




hVB22B 


u2 


-wz4 


VL: 


FR3/SEQ ID NO: 


304 




hVB22B 


u2 


-wz4 


VL: 


CDR3/SEQ ID NO: 


95 


30 


hVB22B 


u2 


-wz4 


VL: 


FR4/SEQ ID NO: 


305. 



In the present invention, SEQ ID NOs of the CDRs and FRs in the 
hVB22B q-wz5 VH sequence are shown below: 
hVB22B q-wz5 VH : FRl/SEQ ID NO: 298 
hVB22B q-wz5 VH : CDR1/SEQ ID NO: 36 
35 hVB22B q-wz5 VH : FR2/SEQ ID NO: 299 
hVB22B q-wz5 VH : CDR2/SEQ ID NO: 37 



23 



hVB22B q-wz5 VH : FR3/SEQ ID NO: 306 
hVB22B q-wz5 VH : CDR3/SEQ ID NO: 38 
hVB22B q-wz5 VH: FR4/SEQ ID NO: 301. 

In the present invention, SEQ ID NOs of the CDRs and FRs in the 
hVB22B q-wz5 VL sequence are shown below: 
hVB22B q-wz5 VL: FRl/SEQ ID NO: 302 
hVB22B q-wz5 VL: CDR1/SEQ ID NO: 93 
hVB22B q-wz5 VL: FR2/SEQ ID NO: 307 
hVB22B q-wz5 VL: CDR2/SEQ ID NO: 94 
hVB22B q-wz5 VL: FR3/SEQ ID NO: 308 
hVB22B q-wz5 VL: CDR3/SEQ ID NO: 95 
hVB22B q-wz5 VL: FR4/SEQ ID NO: 305'. 

The CDRs and FRs in the hVB22B p-z sequence, hVB22B g-e sequence, 
hVB22B e sequence, hVB22B u2-wz4 sequence, and hVB22B q-wz5 sequence 

are shown in Fig. 18. 

In other embodiments, preferred humanized antibodies of the 

present invention include: 

humanized antibodies comprising a heavy chain variable region which 
has FRl, 2, 3, and 4 comprising amino acid sequences of any one of 
(1) to (5) indicated below: 

(1) SEQ ID NOs: 230, 232, 234, and 236 (hVB22B p-z: H chain FRl, 2, 
3, and 4) , 

(2) SEQ ID NOs: 265, 267, 269, and 271 (hVB22B g-e: H chain FRl, 2, 
3, and 4) , 

(3) SEQ ID NOs: 279, 281, 283, and 285 (hVB22B e: H chain FRl, 2, 
3, and 4) , 

(4) SEQ ID NOs: 298, 299, 300, and 301 (hVB22B u2-wz4: H chain FRl, 
2, 3, and 4) , and 

(5) SEQ ID NOs : 298, 299, 306, and 301 (hVB22B q-wz5: H chain FRl, 

2, 3, and 4) ; 

humanized antibodies comprising a light chain variable region which 
has FRl, 2, 3, and 4 comprising amino acid sequences of any one of 
(1) to (4) listed below: 

(1) SEQ ID NOs : 239, 241, 243, and 245 (hVB22B p-z: L chain FRl, 2, 

3, and 4) , 

(2) SEQ ID NOs: 272, 274, 276, and 278 (hVB22B g-e or hVB22.B e: L 



24 



chain FR1, 2, 3 r and 4), 

(3) SEQ ID NOs: 302, 303, 304, and 305 (hVB22B u2-we4 : L chain FR1, 
2 , 3 , and 4 ) , and 

(4) SEQ ID NOs: 302, 307, 308, and 305 (hVB22B q-wz5: L chain FR1, 
5 2, 3, and 4); 

humanized antibodies comprising a heavy chain variable region which 
has CDRl, 2 and 3 comprising amino acid sequences according to the 
SEQ ID NOs listed below: 

SEQ ID NOs: 36, 37, and 38 (hVB22B p-z, hVB22B g-e, hVB22B e, hVB22B 
10 u2-wz4, or hVB22B q~wz5: H chain CDRl, 2, and 3); and 

humanized antibodies comprising a light chain variable region which 
has CDRl, 2 and 3 comprising amino acid sequences according to the 
SEQ ID NOs listed below: 

SEQ ID NOs: 93, 94, and 95 (hVB22B p-z hVB22B g-e, hVB22B e, hVB22B 
15 u2-wz4, or hVB22B q-wz5: L chain CDRl, 2, and 3). 

In yet another preferred embodiment, preferred humanized 
antibodies of the present invention include: 

humanized antibodies comprising heavy chain and light chain variable 
regions of any one of (1) to (5) indicated below. 

20 (1) a heavy chain variable region which comprises FR1, 2, 3, and 4 
comprising the amino acid sequences of SEQ ID NOs: 230, 232, 234, 
and 236, respectively, and a light chain variable region which 
comprises FR1, 2, 3, and 4 comprising the amino acid sequences of 
SEQ ID NOs: 239, 241, 243, and 245, respectively; 

25 (2) a heavy chain variable region which comprises FR1 , 2, 3, and 4 
comprising the amino acid sequences of SEQ ID NOs: 265, 267, 269, 
and 271, respectively, and a light chain variable region which 
comprises FR1, 2, 3, and 4 comprising the amino acid sequences of- 
SEQ ID NOs: 272, 274, 276, and 278, respectively; 

30 (3) a heavy chain variable region which comprises FR1, 2, 3 and 4 
comprising the amino acid sequences of SEQ ID NOs: 279, 281, 283, 
and 285, respectively, and a light chain variable region which 
comprises FR1, 2, 3, and 4 comprising the amino acid sequences of 
SEQ ID NOs: 272, 274, 276, and 278, respectively; 

35 (4) a heavy chain variable region which comprises FR1, 2, 3, and 4 
comprising the amino acid sequences of SEQ ID NOs: 298, 299, 300, 



25 



and 301, and a light chain variable region which comprises FR1, 2, 
3, and 4 comprising the amino acid sequences of SEQ ID NOs: 302, 303, 
304, and 305, respectively; 

(5) a heavy chain variable region which comprises FR1, 2, 3, and 4 
comprising the amino acid sequences of SEQ ID NOs: 298, 299, 306, 
and 301, respectively, and a light chain variable region which 
comprises FR1, 2, 3, and 4 comprising the amino acid, sequences of 
SEQ ID NOs: 302, 307, 308, and 305, respectively; and 
humanized antibodies comprising heavy chain and light chain variable 
regions described below: 

a heavy chain variable region which comprises CDR1, 2, and 3 comprising 
the amino. acid sequences of SEQ ID NOs: 36, 37, and 38, respectively, 
and a light chain variable region which comprises CDR1, 2, and 3 
comprising the amino acid sequences of SEQ ID NOs: 93, 94, and 95, 
respectively. 

Chimeric antibodies and humanized antibodies exhibit lower 
antigenicity in the human body, and thus are expected to be useful 
when administered to humans for therapeutic purposes. 

In one embodiment, the preferred antibodies of the present 
invention include antibodies that bind to soluble Mpl . The term 
"soluble Mpl" herein refers to Mpl molecules excluding those expressed 
on the cell membrane. A specific example of a soluble Mpl is an Mpl 
lacking the entire or a portion of the transmembrane domain. The 
transmembrane domain of human Mpl corresponds to amino acids 492-513 
in SEQ ID NO: 123. 

An antibody that binds to soluble recombinant Mpl can be used 
in detailed epitope analysis and kinetic analysis of receptor-ligand 
binding, as well as for assessing the blood concentration and dynamic 
behavior of the antibody in in vivo tests. 

In one embodiment, the preferred antibodies of the present 
invention include antibodies having binding activity against both 
human and monkey Mpl . The present invention also provides antibodies 
having agonistic activity to human Mpl and monkey Mpl. Antibodies 
having agonistic activity to both human and monkey Mpl are expected 
to be highly useful since the dynamic behavior and in vivo effects 
of the antibody, which are generally difficult to determine in human 



I 



26 



I 



body, can be examined with monkeys. 

Such antibodies may also have binding activity or agonistic 
activity against Mpl from animals other than humans and monkeys (for 
example, mice) . 

5 In addition, the antibodies of the present invention include 

antibodies with TPO agonistic activity (agonistic activity against 
Mpl) of EC50 = 100 nM or lower, preferably EC50 =. 30 nM or lower, 
more preferably EC50 = 10 nM or lower. 

The agonistic activity can be determined by methods known to 

10 those skilled in the art, for example, by the method described below. 
The sequences for human Mpl (Palacios et al ., Cell 41: 727-734, (1985); 
GenBank Accession NO. NM_005373) , cynomolgus monkey Mpl (the 
nucleotide sequence and amino acid sequence are shown in SEQ ID NO: 
1 64 and SEQ ID NO: 165, respectively), and mouse Mpl (GenBank Access ion 

15 NO. NM_010823) are already known. 

In addition, the present invention includes antibodies whose 
binding activities to soluble Mpl are KD = 10" 6 M or lower, preferably 
KD = 10" 7 M or lower. 

In the present invention, whether the binding activity of an 

20 antibody to soluble recombinant Mpl is KD = 10" 6 M or lower can be 
determined by methods known to those skilled in the art. For example, 
the activity can be determined using surface plasmon resonance with 
Biacore. Specifically, soluble MPL-Fc protein , soluble MPL protein, 
or epitope peptides recognized by antibodies are immobilized onto 

25 sensor chips. Reaction rate constant can be determined by assessing 
the interaction between the antibody and the soluble Mpl-Fc protein, 
the soluble Mpl protein, or the epitope peptide recognized by the 
antibody. The proteins to be immobilized on chips are not limited 
in particular, and include, for example, MG10 (from Gln213 to 

30 Ala231)-GST fusion protein and Mpl-IgG Fc fusion protein described 
in the Examples. Since the antibodies are divalent and have two 
antigen-binding sites, the binding activities of these antibodies 
may be determined as those for the monovalent or divalent antibodies, 
or for mixtures of both. Any of these can be used in the present 

35 invention. 

The binding activity can be evaluated by ELISA (enzyme-linked 



27 



immunosorbent assays), EIA (enzyme immunoassays), RIA (radio 
immunoassays), or fluorescent antibody techniques. For example, in 
enzyme immunoassays, a sample containing a test antibody, such as 
purified antibody or culture supernatant of cells producing the test 
5 antibody, is added to a plate coated with an antigen to which the 
test antibody can bind. After incubating the plate with a secondary 
antibody labeled with an enzyme such as alkaline phosphatase, the 
plate is washed and an enzyme substrate such as p-nitrophenyl 
phosphate is added. The antigen-binding activity can then be 

10 evaluated by determining the absorbance. 

There is no specific limitation as to the upper limit of the 
binding activity; for example, the upper limit may be set within a 
technically feasible range by those skilled in the art. However, the 
technically feasible range may expand with the advancement of 

15 technology. 

In an embodiment, the preferred antibodies of the present 
invention include antibodies recognizing epitopes that are recognized 
by any one of the antibodies indicated in (I) to (XII) below. The 
antibody of any one of (I) to (XII) is preferably a minibody. 

20 

(1) Antibody comprising a VH that has CDR1, 2, and 3 comprising the 
amino acid sequences according to SEQ ID NOs in any one of (1) to 

(17) indicated below (name of each antibody and the H chain CDR 
contained in the antibody are indicated inside the parentheses) : 

25 (1) SEQ ID NOs: 3, 4, and 5 (VA7 : H chain CDRl, 2, and 3), 

(2) SEQ ID NOs: 6, 7, and 8 (VA130 or VB17B: H chain CDRl, 2, and 
3) , 

(3) SEQ ID NOs: 9, 10, and 11 (VA259: H chain CDRl, 2, and 3), 

(4) SEQ ID NOs: 15, 16, and 17 (VB12B: H chain CDRl, 2, and 3), 
30 (5) SEQ ID NOs: 18, 19, and 20 (VB140: H chain CDRl, 2, and 3), 

(6) SEQ ID NOs: 21, 22, and 23 (VB33: H chain CDRl, 2, and 3), 

(7) SEQ ID NOs: 24, 25, and 26 (VB45B: H chain CDRl, 2, and 3), 

(8) SEQ ID NOs: 27, 28, and 29 (VB8B : H chain CDRl, 2, and 3), 

(9) SEQ ID NOs: 30, 31, and 32 (VB115: H chain CDRl, 2, and 3), 
35 (10) SEQ ID NOs: 33, 34, and 35 (VB14B: H chain CDRl, 2, and 3), 

(11) SEQ ID NOs: 36, 37, and 38 (VB22B, VB4B, hVB22B p-Z, hVB22B g-e, 



28 



hVB22B e, hVB22B u2-wz4 or hVB22B q-wz5: H chain CDRl, 2, and 3), 

(12) SEQ ID NOs: 39, 40, and 41 (VB16: H chain CDRl, 2, and 3), 

(13) SEQ ID NOs: 42, 43, and 44 (VB157: H chain CDRl, 2, and 3), 

(14) SEQ ID NOs: 48, 49, and 50 (VB51: H chain CDRl, 2, and 3), 

(15) SEQ ID NOs: 51, 52, and 53 (AB317: H chain CDRl, 2, and 3), 

(16) SEQ ID NOs: 54, 55, and 56 (AB324: H chain CDRl, 2, and 3), 

(17) SEQ ID NOs: 57, 58, and 59 (TA136: H chain CDRl, 2, and 3). 

(II) Antibody comprising a VL which has CDRl, 2, and 3 comprising 
the amino acid sequences according to SEQ ID NOs in any one of (1) 
to (10) indicated below (name of each antibody and the L chain CDR 
in the antibody are indicated inside the parentheses) : 

(1) SEQ ID NOs: 60, 61, and 62 (VA7: L chain CDRl, 2, and 3), 

(2) SEQ ID NOs: 63, 64, and 65 (VA130, VA259, VB17B, VB12B, VB140, 
VB45B, VB115, VB14B or VB51: L chain CDRl, 2, and 3), 

(3) SEQ ID NOs: 78, 79, and 80 (VB33 or VB157: L chain CDRl, 2, and 
3) , 

(4) SEQ ID NOs: 84, 85, and 86 (VB8B: L chain CDRl, 2, and 3), 

(5) SEQ ID NOs: 93, 94, and 95 (VB22B, hVB22B p-z, hVB22B g-e, hVB22B 
e, hVB22B u2-wz4 or hVB22B q-wz5: L chain CDRl, 2, and 3), 

(6) SEQ ID NOs: 96, 97, and 98 (VB16: L chain CDRl, 2, and 3), 

(7) SEQ ID NOs: 102, 103, and 104 (VB4B: L chain CDRl, 2, and 3), 

(8) SEQ ID NOs: 108, 109, and 110 (AB317: L chain CDRl, 2, and 3), 

(9) SEQ ID NOs: 111, 112, and 113 (AB324: L chain CDRl, 2, and 3), 

(10) SEQ ID NOs: 114, 115, and 116 (TA136: L chain CDRl, 2, and 3) . 



(Ill) Antibody comprising a VH that comprises an amino acid sequence 



of 


the SEQ 


ID 


NO in 


any one of 


(1) 


SEQ 


ID 


NO: 


124 


(VA7 : VH) , 


(2) 


SEQ 


ID 


NO: 


126 


(VA130: VH)., 


(3) 


SEQ 


ID 


NO: 


128. 


(VA259: VH) , 


(4) 


SEQ 


ID 


NO: 


130 


(VB17B: VH) , 


(5) 


SEQ 


ID 


NO: 


132 


(VB12B: VH) , 


(6) 


SEQ 


ID 


NO: 


134 


(VB140: VH) , 


(7) 


SEQ 


ID 


NO: 


136 


(VB33: VH), 


(8) 


SEQ 


ID 


NO: 


138 


(VB45B: VH) , 



29 



(9) SEQ ID NO: 140 (VB8B: VH) , 



(10) 


SEQ 


ID 


NO: 


142 


(VB115: VH) , 


(11) 


SEQ 


ID 


NO: 


144 


(VB14B: VH), 


(12) 


SEQ 


ID 


NO: 


118 


(VB22B: VH) , 


( 13 ) 


SEQ 


ID 


NO: 


146 


(VB16: VH) , 




SEO 


ID 


NO: 


148 


(VB157 : VH) , 


( lo ) 








X *J V 


fVB4B- VH) , 


(16) 


SEQ 


ID 


NO: 


152 


(VB51: VH), 


(11) 


SEQ 


ID 


NO: 


155 


(AB317: VH) , 


(18) 


SEQ 


ID 


NO: 


159 


(AB324: VH) , 


(19) 


SEQ 


ID 


NO: 


162 


(TA136: VH) , 


(20) 


SEQ 


ID 


NO: 


229 


(hVB22B p-z: VH) , 


(21) 


SEQ 


ID 


NO: 


256 


(hVB22B g-e: VH) , 


(22) 


SEQ 


ID 


NO: 


262 


(hVB22B e: VH) , 


(23) 


SEQ 


ID 


NO: 


289 


(hVB22B u2-wz4: VH) , 


(24) 


SEQ 


ID 


NO: 


295 


(hVB22B q-wz5: VH) . 



(IV) Antibody comprising a VL that comprises an amino acid sequence 
of the SEQ ID NO in any one of (1) to (18): 

(1) SEQ ID NO: 125 (VA7 : VL) , 

(2) SEQ ID NO: 127 (VA130, VB17B, VB12B, VB115 or VB14B: VL) , 

(3) SEQ ID NO: 129 (VA259: VL) , 

(4) SEQ ID NO: 135 (VB140 or VB45B: VL) , 

(5) SEQ ID NO: 137 (VB33: VL) , 

(6) SEQ ID NO: 141 (VB8B: VL) , 

(7) SEQ ID NO: 120 ( VB22B : VL) , 

(8) SEQ ID NO: 147 (VB16: VL) , 

(9) SEQ ID NO: 149 (VB157: VL) , 

(10) SEQ ID NO: 151 (VB4B: VL) , 

(11) SEQ ID NO: 153 (VB51: VL) , 

(12) SEQ ID NO: 157 (AB317: VL) , 

(13) SEQ ID NO: 161 (AB324: VL) , 

(14) SEQ ID NO: 163 (TA136: VL) , 

(15) SEQ ID NO: 238 (hVB22B p-z: VL) , 

(16) SEQ ID NO: 258 (hVB22B g-e: VL or hVB22B e: VL) , 

(17) SEQ ID NO: 291 (hVB22B u2-wz4: VL) , 



30 



(18) SEQ ID NO: 297 (hVB22B q-wz5: VL) . 

(V) Antibody comprising a VH and VL according to any one of (1) to 
(18) : 

5 (1) SEQ ID NOs: 3, 4, and 5 (VA7 : H chain CDR1, 2, and 3); SEQ ID 
NOs: 60, 61, and 62 (VA7: L chain CDR1, 2, and 3), 
(2) SEQ ID NOs: 6, 7, and 8 (VA130 or VB17B: H chain CDRl, 2, and 
3), SEQ ID NOs: 63, 64, and 65 (VA130 or VB17B: L chain CDR1, 2, and 
3) , 

10 (3) SEQ ID NOs: 9, 10, and 11 (VA259: H chain CDRl, 2, and 3); SEQ 
ID NOs: 66, 67, and 68 (VA259: L chain CDRl, 2, and 3), 

(4) SEQ ID NOs : 15, 16, and 17 ( VB12B : H chain CDRl, 2, and 3); SEQ 
ID NO: 72, 73, and 74 (VB12B: L chain CDRl, 2, and 3), 

(5) SEQ ID NOs: 18, 19, and 20 (VB140: H chain CDRl, 2, and 3); SEQ 
15 ID NOs: 75, 76, and 77 (VB140: L chain CDRl, 2, and 3), 

(6) SEQ ID NOs: 21, 22, and 23 (VB33: H chain CDRl, 2, and 3); SEQ 
ID NOs: 78, 79, and 80 (VB33: L chain CDRl, 2, and 3), 

(7) SEQ ID NOs : 24, 25, and 26 (VB45B: H chain CDRl, 2, and 3); SEQ 
ID NOs: 81, 82, and 83 (VB45B: L chain CDRl, 2, and 3), 

20 (8) SEQ ID NOs : 27, 23, and 29 (VB8B: H chain CDRl, 2, and 3); SEQ 
ID NOs: 84-, 85, and 86 { VB8B: L chain CDRl, 2, and 3), 

(9) SEQ ID NOs : 30, 31, and 32 (VB115: H chain CDRl, 2, and 3); SEQ 
ID NOs: 87, 88, and 89 (VB115: L chain CDRl, 2, and 3), 

(10) SEQ ID NOs: 33, 34, and 35 (VB14B: H chain CDRl, 2, and 3); SEQ 
25 ID NOs: 90, 91, and 92 (VB14B: L chain CDRl, 2, and 3), 

(11) SEQIDNOs: 36, 37, and 38 (VB22B, hVB22Bp-z, hVB22B g-e, hVB22B 
e, hVB22B u2-wz4 or hVB22B q-wz5: H chain CDRl, 2, and 3); SEQ ID 
NOs: 93, 94, and 95 (VB22B, hVB22B p-z, hVB22B g-e, hVB22B e, hVB22B 
u2-wz4 or hVB22B q-wz5: L chain CDRl, 2, and 3), 

30 (12) SEQ ID NOs: 39, 40, and 41 (VB16: H chain CDRl, 2, and 3); SEQ 
ID NOs: 96, 97, and 98 (VB16: L chain CDRl, 2, and 3), 

(13) SEQ ID NOs: 42, 43, and 44 (VB157: H chain CDRl, 2, and 3); SEQ 
ID NOs: 99, 100, and 101 (VB157: L chain CDRl, 2, and 3), 

(14) SEQ ID NOs: 45, 46, and 47 (VB4B: H chain CDRl, 2, and 3); SEQ 
35 ID NOs: 102, 103, and 104 (VB4B: L chain CDRl, 2, and 3), 

(15) SEQ ID NOs: 48, 49, and 50 (VB51: H chain CDRl, 2, and 3); SEQ 



31 



ID NOs: 105, 106, and 107 (VB51: L chain CDR1, 2, and 3), 

(16) SEQ ID NOs r 51, 52, and 53 (AB317: H chain CDR1, 2, and 3); SEQ 
ID NOs : 108, 109, and 110 (AB317: L chain CDR1, 2, and 3), 

(17) SEQ ID NOs: 54, 55, and 56 (AB324: H chain CDR1, 2, and 3); SEQ 
ID NOs : 111, 112, and 113 (AB324 : L chain CDR1, 2, and 3), 

(18) SEQ ID NOs: 57, 58, and 59 (TA136: H chain CDR1 , 2, and 3); SEQ 
ID NOs : 114, 115, and 116 (TA136: L chain CDR1, 2, and 3). 

(VI) Antibody comprising a VH and a VL that comprise the amino acid 
sequences according to SEQ ID NOs in any one of (1) to (24) indicated 
below: 

(1) SEQ ID NO: 124 (VA7 : VH) , SEQ ID NO: 125 (VA7 : VL) , 

(2) SEQ ID NO: 126 (VA130: VH) , SEQ ID NO: 127 (VA130: VL) , 

(3) SEQ ID NO: 128 (VA259: VH) , SEQ ID NO: 129 (VA259: VL) , 

(4) SEQ ID NO: 130 (VB17B: VH) , SEQ ID NO: 127 (VB17B: VL) , 

(5) SEQ ID NO: 132 (VB12B: VH ) , SEQ ID NO: 127 (VB12B: VL) , 

(6) SEQ ID NO: 134 (VB140: VH) , SEQ ID NO: 135 (VB140: VL) , 

(7) SEQ ID NO: 136 (VB33: VH) , SEQ ID NO: 137 (VB33: VL), 



(8) SEQ ID NO: 138 (VB45B: VH) , SEQ ID NO: 135 (VB45B: VL) , 

(9) SEQ ID NO: 140 (VB8B: VH) , SEQ ID NO: 141 (VB8B: VL) , 



(10) 


SEQ 


ID 


NO: 


142 


(VB115: 


VH) , 


SEQ ID NO 


127 


(VB115: VL), 


(11) 


SEQ 


ID 


NO: 


144 


(VB14B: 


VH) , 


SEQ ID NO 


: 127 


(VB14B: VL) , 


(12) 


SEQ 


ID 


NO: 


118 


(VB22B : 


VH) , 


SEQ ID NO 


: 12 0 


(VB22B: VL), 


(13) 


SEQ 


ID 


NO: 


146 


(VB16: VH) , 


SEQ ID NO: 


147 


(VB16: VL) , 


(14) 


SEQ 


ID 


NO: 


148 


(VB157: 


VH) , 


SEQ ID NO 


: 149 


(VB157: VL), 


(15) 


SEQ 


ID 


NO: 


150 


(VB4B: VH) , 


SEQ ID NO: 


151 


(VB4B: VL), 


<16) 


SEQ 


ID 


NO: 


152 


(VB51: VH) , 


SEQ ID NO: 


153 


(VB51: VL), 


(17) 


SEQ 


ID 


NO: 


155 


(AB317 : 


VH) , 


SEQ ID NO 


: 157 


(AB317: VL), 


(18) 


SEQ 


ID 


NO: 


159 


(AB324 : 


VH) , 


SEQ ID NO 


: 161 


(AB324: VL) , 


(19) 


SEQ 


ID 


NO: 


162 


(TA136: 


VH) , 


SEQ ID NO 


: 163 


(TA136: VL) , 


(20) 


SEQ 


ID 


NO: 


229 


(hVB22B 


p-z: 


VH), SEQ 


ID NO 


: 238 (hVB22B 


VL) , 




















(21) 


SEQ 


ID 


NO: 


256 


(hVB22B 


g-e: 


VH) , SEQ 


ID NO 


: 258 (hVB22B 


VL) , 




















(22) 


SEQ 


ID 


NO: 


262 


(hVB22B 


e: VH) , SEQ ID 


NO: 258 (hVB22B e: 


(23) 


SEQ ID NO: 289 (hVB22B u2 


-wz4 


. VH) , SEQ ID NO: 


291 (hVB22B u2 



32 



VL) , 

(24) SEQ ID NO: 295 (hVB22B q-wz5 : VH) , SEQ ID NO: 297 (HVB22B q-wz5: 
VL) . 

5 (VII) Antibody comprising the amino acid sequence of SEQ ID NO: 122 
(VB22B: scFv) . 

(VIII) Humanized antibody comprising an amino acid sequence 
according to any one of SEQ ID NO: 2 (hVB22B p-z: sc(Fv)2), SEQ ID 

10 NO: 254 (hVB22B g-e: sc(Fv)2), SEQ ID NO: 260 (hVB22B e: sc(Fv)2), 
SEQ ID NO: 287 (hVB22B u2-wz4 : sc(Fv)2),' and SEQ ID NO: 293 (hVB22B 
q-wz5 : sc (Fv) 2 ) . 

(IX) Antibody comprising a VH which has FR1, 2, 3, and 4 comprising 
15 amino acid sequences according to SEQ ID NOs in any one of (1) to 

(5) indicated below: 

(1) SEQ ID NOs: 230, 232, 234, and 236 (hVB22B p-z: H chain FR1, 2, 
3, and 4 ) , 

(2) SEQ ID NOs: 265, 267, 269, and 271 (hVB22B g-e: H chain FR1, 2, 
20 3, and 4 ) , 

(3) SEQ ID NOs: 279, 281, 283, and 285 (hVB22B e: H chain FR1, 2, 
3, and 4 ) , 

(4) SEQ ID NOs: 298, 299, 300, and 301 (hVB22B u2-wz4 : H chain FR1, 
2, 3, and 4) , 

25 (5) SEQ ID NOs: 298, 299, 306, and 301 (hVB22B q-wz5: H chain FR1 , 

2, 3, and 4 ) . 

(X) Antibody comprising a VL which has FR1, 2, 3 and 4 comprising 
amino acid sequences according to SEQ ID NOs in any one of (1) to 

30 (4) indicated below: 

(1) SEQ ID NOs: 239, 241, 243, and 245 (hVB22B p-z: L chain FR1 , 2, 

3, and 4 ) , 

(2) SEQ ID NOs: 272, 274, 276, and 278 (hVB22B g-e or hVB22B e: L 
chain FR1 , 2, 3, and 4), 

35 (3) SEQ ID NOs: 302, 303, 304, and 305 (hVB22B u2-wz4: L chain FR1, 
2, 3, and 4) , 



33 



(4) SEQ ID NOs: 302, 307, 308, and 305 (hVB22B q-wz5: L chain FR1, 
2, 3, and 4) . 

(XI) Antibody comprising VH and VL according to any one of (1) to 
5 (5) indicated below: 

(1) VH having FR1, 2, 3, and 4 comprising the amino acid sequences 
of SEQ ID NOs: 230, 232, 234, and 236, respectively, and VL having 
FR1, 2, 3, and 4 comprising the amino acid sequences of SEQ ID NOs: 
239, 241, 243, and 245, respectively; 
10 (2) VH having FR1, 2, 3, and 4 comprising the amino acid sequences 
of SEQ ID NOs: 265, 267, 269, and 271, respectively, and VL having 
FR1, 2, 3, and 4 comprising the amino acid sequences of SEQ ID NOs: 
272, 274, 276, and 278, respectively; 

(3) VH having FR1, 2, 3, and 4 comprising the amino acid sequences 
15 of SEQ ID NOs: 279, 281, 283, and 265, respectively, and VL having 

FR1, 2, 3, and 4 comprising the amino acid sequences of SEQ ID NOs: 
272, 274, 276, and 278, respectively; 

(4) VH having FR1, 2, 3, and 4 comprising the amino acid sequences 
of SEQ ID NOs: 298, 299, 300, and 301, respectively, and VL having 

20 FR1, 2, 3, and 4 comprising the amino acid sequences of SEQ ID NOs: 
302, 303, 304, and 305, respectively; 

(5) VH having FR1, 2, 3, and 4 comprising the amino acid sequences 
of SEQ ID NOs: 298, 299, 306, and 301, respectively, and VL having 
FR1, 2, 3, and 4 comprising the amino acid sequences of SEQ ID NOs: 

25 302, 307, 308, and 305, respectively. 

(XII) Antibody comprising the amino acid sequence of SEQ ID NO: 264 
(VB22B: sc (Fv) 2) . 

An antibody comprising an amino acid sequence of any one of (I) 
30 to (XII) indicated above, in which one or more amino acids have been 
substituted, deleted, added, and/or inserted, wherein the antibody 
has activity equivalent to that of the antibody of any one of (I) 
to (XII) . 

Herein, the phrase "functionally equivalent" means that an 
35 antibody of interest has a biological or biochemical activity 
comparable to that of an antibody of the present invention. Such 



34 



activities include, for example, binding activities and agonistic 
activities . 

Methods for preparing polypeptides functionally equivalent to 
a certain polypeptide are well known to those skilled in the art, 

5 and include methods of introducing mutations into polypeptides. For 
example, those skilled in the art'can prepare an antibody functionally 
equivalent to the antibodies of the present invention by introducing 
appropriate mutations into the antibody using site-directed 
mutagenesis (Hashimoto-Gotoh, T. et al. Gene 152, 271-275, (1995); 
10 Zoller, MJ, and Smith, M. Methods Enzymol. 100, 468-500, (1983); 
Kramer, W. et al., Nucleic Acids Res . 12, 9441-9456, (1984); Kramer, 
W. and Fritz HJ, Methods Enzymol. 154, 350-367, (1987); Kunkel, TA, 
Proc. Natl. Acad. Sci . USA. 82, 488-492, (1985); Kunkel, Methods 
Enzymol. 85, 2763-2766, (1988)), or such . Amino acid mutations may 

15 occur naturally. Thus, the present invention also comprises 
antibodies functionally equivalent to the antibodies of the present 
invention and comprising the amino acid sequences of these antibodies, 
in which one or more amino acids is mutated. In such mutants, the 
number of amino acids that may be mutated is not particularly 

20 restricted, so long as the activity is maintained. Generally, the 
number of amino acids that are mutated is 50 amino acids or less, 
preferably 30 or less, more preferably 10 or less (for example, five 
amino acids or less) . Likewise, the site of mutation is not 
particularly restricted, so long as the mutation does not result in 

25 the disruption of activity. 

Amino acid mutations may be made at one or more predicted, 
preferably nonessential, amino acid residues. A "nonessential" 
amino acid residue is a residue that can be altered from the wild-type 
sequence of a protein without altering the biological activity, 

30 whereas an "essential" amino acid residue is required for biological 
activity. An amino acid is preferably substituted for a different 
amino acid(s) that allows the properties of the amino acid side-chain 
to be conserved. Accordingly, throughout the present application, 
a "conservative amino acid substitution" means a replacement of an 

35 amino acid residue belonging to one of the following groups with 
another amino acid in the same group having a chemically similar side 



35 



chain. Groups of amino acid residues having similar side chains have 
been defined in the art. Examples of amino acid side chain properties 
are: hydrophobic amino acids (A, I, L, M, F, P, W, Y, and V) , hydrophilic 
amino acids ( R, D, N, C, E, Q, G, H, K, S, and T) , amino acids comprising 
5 the following side chains: aliphatic side chains (G, A, V, L, I, and 
P) ; hydroxyl-containing side chains (S, T, and Y) ; sulfur-containing 
side chains (C and M) ; carboxylic acid- and amide-containing side 
chains (D, N, E, and Q) ; basic side chains (R, K, and H) ; aromatic 
ring-containing side chains (H, F, Y , and W) (amino acids are 
10 represented by one-letter codes in parentheses) . 

A polypeptide comprising a modified amino acid sequence, in 
which one or more amino acid residues is deleted, added, and/or 
replaced with other amino acids, is known to retain its original 
biological activity (Mark, D. F. et al., Proc. Natl. Acad. Sci. USA 
15 81, 5662-5666 (1984); Zoller, M. J. & Smith, M. Nucleic Acids Research 
10, 6487-6500 (1982); Wang, A. et al., Science 224, 1431-1433; 
Dalbadie-McFarland, G. et al., Proc. Natl. Acad. Sci. USA "79, 
6409-6413 (1982) ) . 

Fusion proteins containing antibodies that comprise the amino 
20 acid sequence of an antibody of the present invention, in which two 
or more amino acid residues have been added, are included in the 
present invention. The fusion protein results from a fusion between 
one of the above antibodies and a second peptide or protein, and is 
included in the present invention. The fusion protein can be prepared 
25 by ligating a polynucleotide encoding an antibody of the present 
invention and a polynucleotide encoding a second peptide or 
polypeptide in frame, inserting this into an expression vector, and 
expressing the fusion construct in a host. Some techniques known to 
those skilled in the art are available for this purpose. The partner 
30 peptide or polypeptide to be fused with an antibody of the present 
invention may be a known peptide, for example, FLAG (Hopp, T. P. et 
al., BioTechnology 6, 1204-1210 (1988)), 6x His consisting of six 
His (histidine) residues, lOx His, influenza hemagglutinin (HA) , 
human c-myc fragment, VSV-GP fragment, pl8HIV fragment, T7-tag, 
35 HSV-tag, E-tag, SV40 T antigen fragment, lck tag, a-tubulin fragment, 
B-tag, Protein C fragment. Other partner polypeptides to be fused 



36 



with the antibodies of the present invention include, for example, 
GST (glutathione-S-transferase) , HA (influenza hemagglutinin), 
immunoglobulin constant region, p-galactosidase, and MBP 
(maltose-binding protein) . A polynucleotide encoding one of these 
5 commercially available peptides or polypeptides can be fused with 
a polynucleotide encoding an antibody of the present invention. The 
fusion polypeptide can be prepared by expressing the fusion construct . 

As described below, the antibodies of the present invention may 
differ in amino acid sequence, molecular weight, isoelectric point, 

10 presence/absence of sugar chains, and conformation depending on the 
cell or host producing the antibody, or purification method . However, 
a resulting antibody is included in the present invention, as long 
as it is functionally equivalent to an antibody of the present 
invention. For example, when an antibody of the present invention 

15 is expressed in prokaryotic cells, for example E. coli, a methionine 
residue is added to the N terminus of the original antibody amino 
acid sequence. Such antibodies are included in the present 
invention . 

An antibody that recognizes an epitope recognized by the 

20 antibody according to any one of (I) to (XII) indicated above is 
expected to have a high agonistic activity. Such antibodies can be 
prepared by methods known to those skilled in the art. The antibody 
can be prepared by, for example, determining the epitope recognized 
by the antibody according to any one of (I) to (XII) by conventional 

25 methods, and using a polypeptide comprising one of the epitope amino 
acid sequences as an immunogen. Alternatively, the antibody can be 
prepared by determining the epitopes of conventionally prepared 
antibodies and selecting an antibody that recognizes the epitope 
recognized by an antibody of any one of (I) to (XII) . 

30 In the present invention, a particularly preferred antibody is 

an antibody that recognizes the epitope recognized by the antibody 
comprising the amino acid sequence of SEQ ID NO: 2. The antibody 
comprising the amino acid sequence of SEQ ID NO: 2 is predicted to 
recognize the region from Glu 26 to Leu 274, preferably the region 

35 from Ala 189 to Gly 245, more preferably the region from Gin 213 to 
Ala 231 of human Mpl . Thus, antibodies recognizing the region of amino 



37 



acids 26 to 274, or amino acids 189 to 245, or amino acids 213 to 
231 of human Mpl are also included in the present invention. 

Antibodies recognizing regions of amino acids 26 to 274, amino 
acids 189 to 245, or amino acids 213 to 231 of the human Mpl amino 
5 acid sequence (SEQ ID NO: 123) can be obtained by methods known to 
those skilled in the art. Such antibodies can be prepared by, for 
example, using a peptide comprising amino acids 26 to 274, amino acids 
189 to 245, or amino acids 213 to 231 of the human Mpl amino acid 
sequence (SEQ ID NO: 123) as an immunogen. Alternatively, such 
10 antibodies can be prepared by determining the epitope of a 
conventionally prepared antibody and selecting an antibody that 
recognizes the same epitope recognized by an antibody of the present 
invention . 

The present invention provides antibodies described above in 
15 (I) to (XII) . In an embodiment of the present invention, a preferred 
antibody is the one shown in (V) , a more preferred antibody is the 
one shown in (VI), and a still more preferred is the one shown in 
(VIII). 

The present invention also provides vectors comprising 

20 polynucleotides encoding the antibodies of the present invention, 
or polynucleotides which hybridize under stringent conditions to the 
polynucleotides of the present invention and encode antibodies having 
activities equivalent to those of the antibodies of the present 
invention. The polynucleotides of the present invention are polymers 

25 comprising multiple bases or base pairs of deoxyribonucleic acids 
(DNA) or ribonucleic acids (RNA) , and are not particularly limited, 
as long as they encode the antibodies of the present invention. They 
may also contain non-natural nucleotides . The polynucleotides of the 
present invention can be used to express antibodies using genetic 

30 engineering techniques. The polynucleotides of this invention can 
also be used as probes in the screening of antibodies functionally 
equivalent to the antibodies of the present invention. Specifically, 
DNAs that hybridize under stringent conditions to a polynucleotide 
encoding an antibody of the present invention, and encode antibodies 

35 having activity equivalent to those of the antibodies of the present 
invention can be obtained by techniques such as hybridization and 



38 



gene amplification (for example, PCR) , using a polynucleotide of the 
present invention or a portion thereof as a probe. Such DNAs are also 
included in the polynucleotides of the present invention. 
Hybridization techniques are well known to those skilled in the art 
5 (Sambrook, J et al . , Molecular Cloning 2nd ed. , 9.47-9.58, Cold Spring 
Harbor Lab. press, 1989). Such hybridization conditions include, for 
example, conditions of low stringency. Examples of conditions of low 
stringency include post-hybridization washing in O.lx SSC and 0.1% 
SDS at 42°C, and preferably in O.lx SSC and 0.1% SDS at 50°c. More 

10 preferable hybridization conditions include those of high stringency. 
Highly stringent conditions include, for example, washing in 5x SSC 
and 0.1% SDS at 65 °C . In these conditions , the higher the temperature , 
the higher the expectation of efficiently obtaining polynucleotides 
with a high homology. However, several factors, such as temperature 

15 and salt concentration, can influence hybridization stringency, and 
those skilled in the art can suitably select these factors to 
accomplish similar stringencies. 

Antibodies that are encoded by polynucleotides obtained by the 
hybridization and gene amplification techniques, and are functionally 

20 equivalent to the antibodies of the present invention generally 
exhibit high homology to the antibodies of the this invention at the 
amino acid level. The antibodies of the present invention include 
antibodies that are functionally equivalent to the antibodies of the 
present invention, and exhibit high amino acid sequence homology to 

25 the antibodies of this invention. The term "high homology" generally 
means identity at the amino acid level of at least 50% or higher, 
preferably 75% or higher, more preferably 85% or higher, still more 
preferably 95% or higher. Polypeptide homology can be determined by 
the algorithm described in the report: Wilbur, W. J. and Lipman, D. 

30 J. Proc. Natl. Acad. Sci. USA 80, 726-730 (1983). 

When E. coli is used as a host, there is no particular limitation 
as to the type of vector of the present invention, as long as the 
vector contains an "ori" responsible for its replication in E. coli 
and a marker gene. The "ori" ensures the amplification and mass 

35 production of the vector in E. coli (for example, JM109, DH5a, HB101, 
and XLlBlue) . The marker gene is used to select the E. coli 



39 



transf ormants (for example, a drug resistance gene selected by an 
appropriate drug such as ampicillin r tetracycline, kanamycin, and 
chloramphenicol) . The vectors include, for example, M13 vectors, pUC 
vectors, pBR322, pBluescript, and pCR-Script . In addition to the 
above vectors, for example, pGEM-T, pDIRECT, and pT7 can also be used 
for the subcloning and excision of cDNAs. 

In particular, expression vectors are useful as vectors of the 
present invention. When an expression vector is expressed, for 
example, in E. coli, it should have the above characteristics in order 
to be amplified in E. coli. Additionally, when E . coli, such as JM109, 
DH5a, HB101, or XLl-Blue are used as the host cell, the vector 
preferably has a promoter, for example, lacZ, promoter (Ward et al. 
(1989) Nature 341:544-546; (1992) FASEB J. 6:2422-2427), araB 
promoter (Better et al . (1988) Science 240 : 1041-1043) , or T7 promoter , 
that allows efficient expression of the desired gene in E. coli. Other 
examples of the vectors include pGEX-5X-l (Pharmacia), "QIAexpress 
system" (QIAGEN) , pEGFP , and pET (where BL21, a strain expressing 
T7 RNA polymerase, is preferably used as the host) . 

Furthermore, the vectors may comprise a signal sequence for 
polypeptide secretion. When producing polypeptides into the 
periplasm of E . coli, the pelB signal sequence (Lei, S. P. et al. 
J. Bacteriol. 169:4379 (1987)) may be used as a signal sequence for 
polypeptide secretion. For example, calcium chloride methods or 
electroporation methods may be used to introduce the vector into a 
host cell. 

In addition to E . coli, expression vectors derived from mammals 
(e.g., P CDNA3 ( Invitrogen) , pEGF-BOS (Nucleic Acids Res. (1990) 
18 (17) : 5322) , pEF, pCDM8), insect cells (e.g., "Bac-to-BAC 
baculovirus expression system" (G1BCO-BRL) , pBacPAK8)., plants (e.g., 
pMHl, pMH2), animal viruses (e.g., pHSV, pMV, pAdexLcw) , retroviruses 
(e.g., pZIPneo) , yeasts (e.g., "Pichia Expression Kit" (Invitrogen), 
pNVll, SP-Q01) , and Bacillus subtilis (e.g., pPL608, pKTH50) may also 
be used as a vector of the present invention. 

In order to express proteins in animal cells such as CHO, COS, 
and NIH3T3 cells, the vector preferably has a promoter necessary for 
expression in such cells, for example, an SV40 promoter (Mulligan 



40 



et al. (1979) Nature 277:108), MMLV-LTR promoter, EFla promoter 
(Mizushima et al. (1990) Nucleic Acids Res. 18:5322), CMV promoter, 
etc.). It is even more preferable that the vector also carries a 
marker gene for selecting transf ormants (for example, a 
drug-resistance gene selected by a drug such as neomycin and G418. 
Examples of vectors with such characteristics include pMAM, pDR2 , 
pBK-RSV, pBK-CMV, pOPRSV, and pOPl3, and such. 

In addition, to stably express a gene and amplify the gene copy 
number in cells, CHO cells that are defective in the nucleic acid 
synthesis pathway are introduced with a vector containing a DHFR gene 
(for example, pCHOI ) to compensate for the defect, and the copy number 
is amplified using methotrexate (MTX) . Alternatively, a COS cell, 
which carries an SV40 T antigen-expressing gene on its chromosome, 
can be transformed with a vector containing the SV40 replication 
origin (for example, pcD) for transient gene expression. The 
replication origin may be derived from polyoma virus, adenovirus, 
bovine papilloma virus (BPV) , and such. Furthermore, to increase the 
gene copy number in host cells, the expression vector may contain, 
as a selection marker, aminoglycoside transferase (APH) gene, 
thymidine kinase (TK) gene, E. coli xanthine guanine phosphoribosyl 
transferase (Ecogpt) gene, dihydrof olate reductase (dhfr) gene, and 
such . 

In the present invention, next, the vector is introduced into 
a host cell. The host cells into which the vector is introduced are 
not particularly limited, for example, E. coli and various animal 
cells are available for this purpose. The host cells may be used, 
for example, as a production system to produce and express the 
antibodies of the present invention. In vitro and in vivo production 
systems are available for polypeptide production systems. 
Production systems that use eukaryotic cells or prokaryotic cells 
are examples of in vitro production systems. 

Eukaryotic cells that can be used are, for example, animal cells, 
plant cells, and fungal cells. Known animal cells include: mammalian 
cells, for example, CHO (J. Exp. Med. (1995)108, 945), COS, 3T3, 
myeloma, BHK (baby hamster kidney), HeLa, Vero, amphibian cells such 
as Xenopus laevis oocytes (Valle, etal. ( 1 981 ) Nature 2 91 , 358-340), 



41 



or insect cells (e.g., Sf9, Sf21, and Tn5) . In the present invention, 
CHO-DG44, CHO-DXB11, C0S7 cells, and BHK cells can be suitably used. 
Among animal cells, CHO cells are particularly favorable for 
large-scale expression. Vectors can be introduced into a host cell 
5 by, for example, calcium phosphate methods, the DEAE-dextran methods , 
methods using cationic liposome DOTAP (Boehringer-Mannheim) , 
electroporation methods, lipofection methods. 

Plant cells include, for example, Nicotiana tabacum-derived 
cells known as a protein production system. Calluses may be cultured 
10 from these cells . Known fungal cells include yeast cells, for example, 
genus Saccharomyces such as Saccharomyces cerevisiae and 
Saccharomyces pombe; and filamentous fungi, for example, genus 
Aspergillus such as Aspergillus niger. 

Bacterial cells can be used in the prokaryotic production 
15 systems. Examples of bacterial cells include E. coli (for example, 
JM109, DH5a, HB101 and such); and Bacillus subtilis. 

Next, the above host cells are cultured. Antibodies can be 
obtained by transforming the cells with a polynucleotide of interest 
and in vitro culturing of these transf ormants . Transf ormants can be 
20 cultured using known methods . For example, DMEM, MEM, RPMI 1640, or 
IMDM may be used as the culture medium for animal cells, and may be 
used with or without serum supplements such as FBS or fetal calf serum 
(FCS) . Serum-free cultures are also acceptable. The preferred pH 
is about 6 to 8 during the course of culturing. Incubation is carried 
25 out typically at a temperature of about 30 to 40°C for about 15 to 
200 hours. Medium is exchanged, aerated, or agitated, as necessary. 

On the other hand, production systems using animal or plant 
hosts may be used as systems for producing polypeptides in vivo. For 
example, a polynucleotide of interest is introduced into an animal 
30 or plant and the polypeptide is produced in the body of the animal 
or plant and then recovered. The "hosts" of the present invention 
includes such animals and plants. 

Animals to be used for the production system include mammals 
or insects. Mammals such as goats, pigs, sheep, mice, and cattle may 
35 be used (Vicki Glaser SPECTRUM Biotechnology Applications (1993)). 
Alternatively, the mammals may be transgenic animals. 



42 



For example, a polynucleotide of interest is prepared as a 
fusion gene with a gene encoding a polypeptide specifically produced 
in milk, such as the goat P-casein gene. DNA fragments containing 
the fusion gene are injected into goat embryos, which are then 
5 introduced back to f emale goats . The desired antibody can be obtained 
from milk produced by the transgenic goats, which are born from the 
goats that received the embryos, or from their offspring. 
Appropriate hormones may be administered to increase the volume of 
milk containing the antibody produced by the transgenic goats (Ebert, 

10 K.M. et al., Bio/Technology 12, 699-702 (1994)). 

Insects, such as silkworms, may also be used. Baculoviruses 
carrying a polynucleotide encoding an antibody of interest can be 
used to infect silkworms, and the antibody of interest can be obtained 
from the body fluids (Susumu, M. et al., Nature 315, 592-594 (1985)). 

15 Plants used in the production system include, for example, 

tobacco. When tobacco is used, a polynucleotide encoding an antibody 
of interest is inserted into a plant expression vector, for example, 
pMON 530, and then the vector is introduced into a bacterium, such 
as Agrobacter ium tumef aciens . The bacteria are then used to infect 

20 tobacco such as Nicotiana tabacum, and the desired antibodies can 
be recovered from the leaves (Julian K. -C . Ma et al . , Eur . J. Immunol . 
24, 131-138 (1994) ) . 

The resulting antibody may be isolated from the inside or 
outside (such as the medium) of host cells, and purified as a 

25 substantially pure and homogenous antibody. Methods are not limited 
to any specific method and any standard method for isolating and 
purifying antibodies may be used. Polypeptides may be isolated and 
purified, by selecting an appropriate combination of, for example, 
chromatographic columns, filtration, ultrafiltration, salting out, 

30 solvent precipitation, solvent extraction, distillation, 
immunoprecipitation, SDS-polyacrylamide gel electrophoresis, 
isoelectric focusing, dialysis, recrystallization, and others. The 
term "substantially pure" as used herein in reference to a given 
polypeptide means that the polypeptide is substantially free from 

35 contaminants such as other biological macromolecules , culture media 
(if recombinantly produced), or chemical precursors (if chemically 



43 



synthesized). The substantially pure polypeptide is at least 75%, 
preferably at least about 80%, more preferably at least about 85, 
90, 95, or 99% pure by dry weight. Purity can be measured by any 
appropriate standard method, for example by a chromatography method, 
5 polyacrylamide gel electrophoresis, or HPLC analysis. 

Chromatographies include, for example, affinity 
chromatographies, ion exchange chromatographies, hydrophobic 
chromatographies, gel filtrations, reverse-phase chromatographies, 
and adsorption chromatographies (Strategies for Protein Purification 

10 and Characterization: A Laboratory Course Manual . Ed Daniel R. Marshal 
et al., Cold Spring Harbor Laboratory Press, 1996). These 
chromatographies can be carried out using liquid phase 
chromatographies such as HPLC and FPLC. Examples of the affinity 
chromatography columns include protein A columns and protein G columns . 

15 Examples of the proteins A columns include Hyper D, POROS, and 
Sepharose F. F. (Pharmacia) . 

An antibody can be modified freely and peptide portions deleted 
by treating the antibody with an appropriate protein modifying enzyme 
before or after antibody purification. Such protein modifying 

20 enzymes include, for example, trypsins, chymotrypsins, lysyl 
endopeptidases, protein kinases, and glucosidases . 

Antibodies that bind to Mpl can be prepared by methods known 
to those skilled in the art. 

For example, monoclonal antibody-producing hybridomas can be 

25 essentially generated by known technologies as follows: immunizing 
animals with Mpl proteins or Mpl-expressing cells as sensitized 
antigens using conventional immunological methods; fusing the 
obtained immunocytes with known parental cells by conventional cell 
fusion methods; and screening for monoclonal antibody-producing cells 

30 by conventional methods. 

Specifically, monoclonal antibodies can be prepared by the 
method below. 

First, Mpl protein, which is used as a sensitized antigen for 
preparing antibodies, is prepared by expressing the Mpl gene/amino 
35 acid sequence (GenBank accession number: NM_005373). More 
specifically, the gene sequence encoding Mpl is inserted into a known 



44 



expression vector, which is then transf ected into an appropriate host 
cell. The subject human Mpl protein is purified from the host cell 
or culture supernatant using known methods. 

The purified Mpl protein is then used as a sensitized antigen. 
Alternatively, a partial Mpl peptide may be used as a sensitized 
antigen. In this case, the partial peptide can also be chemically 
synthesized based on the amino acid sequence of human Mpl. 

The epitopes of Mpl molecule that are recognized by an anti-Mpl 
antibody of the present invention are not limited to a particular 
epitope, and may be any epitope on the Mpl molecule. Thus, any 
fragment can be used as an antigen for preparing anti-Mpl antibodies 
of the present invention, as long as the fragment comprises an epitope 
of the Mpl molecule. 

There is no limitation as to the type of mammalian species to 
be immunized with the sensitized antigen. However, a mammal is 
preferably selected based on its compatibility with the parental cell 
to be used in cell fusion. Generally, rodents (for example, mice, 
rats, and hamsters), rabbits, and monkeys can be used. 

Animals can be immunized with a sensitized antigen by known 
methods such as a routine method of injecting a sensitized antigen 
into a mammal intraper itoneally or subcutaneously . Specifically, 
the sensitized antigen is diluted appropriately with 
phosphate-buffered saline (PBS), physiological saline and such, and 
then suspended. An adequate amount of a conventional adjuvant, for 
example, Freund' s complete adjuvant, is mixed with the suspension, 
as necessary. An emulsion is then prepared for administering to a 
mammal several times over a 4- to 21-day interval. An appropriate 
carrier may be used for the sensitized antigen in immunization. 

A mammal is immunized as described above. After a titer 
increase of target antibody in the serum is confirmed, immunocytes 
are collected from the mammal and then subjected to cell fusion. 
Spleen cells are the preferred immunocytes. 

Mammalian myeloma cells are used as the parental cells to be 
fused with the above immunocytes . Preferable myeloma cells to be used 
include various known cell lines, for example, P3 ( P3x63Ag8 . 653 ) 
(Kearney JF, et al., J. Immnol. 123, 1548-1550 (1979)), P3x63Ag8U.l 



45 



(Yelton DE, et al., Current Topics in Microbiology and Immunology 
81, 1-7 (1978)), NS-1 (Kohler, G. and Milstein, C. Eur. J. Immunol. 
6, 511-519 (1976)), MPC-11 (Margulies, D. H. et al., Cell 8, 405-415 
(1976)), SP2/0 (Shulman, M. et al., Nature 276, 269-270 (1978)), FO 
5 (deSt. Groth, S. F. et al., J. Immunol. Methods 35, 1-21 (1980)), 
S194 (Trowbridge, I. S., J. Exp. Med. 148, 313-323 (1978)), and R210 
(Galfre, G. et al., Nature 277, 131-133 (1979)). 

Cell fusions between the immunocytes and the myeloma cells as 
described above can be essentially carried out using known methods, 

10 for example, a method by Kohler and Milstein (Kohler, G. and Milstein, 
C, Methods Enzymol. 73, 3-46 (1981)). 

More specifically, the above-described cell fusions are carried 
out, for example, in a conventional culture medium in the presence 
of a cell fusion-promoting agent. The fusion-promoting agents 

15 include, for example, polyethylene glycol (PEG) and Sendai virus (HVJ) . 
If required, an auxiliary substance such as dimethyl sulfoxide may 
also be added to improve fusion efficiency. 

The ratio of immunocytes to myeloma cells may be determined at 
one's own discretion, preferably, for. example, one myeloma cell for 

20 every one to ten immunocytes. Culture media to be used for the above 
cell fusions include, for example, media that are suitable for the 
growth of the above myeloma cell lines, such as RPMI 1640 media and 
MEM media, and other conventional culture media used for this type 
of cell culture. In addition, serum supplements such as fetal calf 

25 serum (FCS) may also be used in combination. 

Cell fusion is carried out as follows. As described above, 
predetermined amounts of immunocytes and myeloma cells are mixed well 
in the culture medium. PEG solution (for example, mean molecular 

weight of about 1, 000-6, 000) pre-heated to 37°C is added to the cell 
30 suspension typically at a concentration of 30% to 60% (w/v) , and mixed 
to produce fused cells (hybridomas) . Then, an appropriate culture 
medium is successively added to the mixture, and the sample is 
centrifuged to remove supernatant. This treatment is repeated 
several times to remove the unwanted cell fusion-promoting agent and 
35 others that are unfavorable to hybridoma growth. 

Screening of the resulting hybridomas can be carried out by 



46 



culturing them in a conventional selective medium, for example, 
hypoxanthine, aminopterin, and thymidine (HAT) medium. Culturing in 
the above-descried HAT medium is continued for a period long enough 
{typically, for several days to several weeks) to kill cells 
5 (non-fused cells) other than the desired hybridomas. Then, 
hybridomas are screened for single-cell clones capable of producing 
the target antibody by conventional limiting dilution methods. 

In addition to the method for preparing the above-descried 
hybridomas by immunizing non-human animals with antigens, preferred 

10 human antibodies having binding activity to Mpl can also be obtained 
by: sensitizing human lymphocytes with Mpl in vitro; and fusing the 
sensitized lymphocytes with human myeloma cells capable of dividing 
permanently (see, Examined Published Japanese Patent Application No. 
(JP-B) Hei 1-59878). Alternatively, it is possible to obtain human 

15 antibodies against Mpl from immortalized cells producing anti-Mpl 
antibodies. In this method, the cells producing anti-Mpl antibodies 
are prepared by administering Mpl as an antigen to transgenic animals 
comprising a repertoire of the entire human antibody genes (see, WO 
94/25585, WO 93/12227, WO 92/03918, and WO 94/02602). 

20 ■ The monoclonal antibody-producing hybridomas thus prepared can 

be passaged in a conventional culture medium, and stored in liquid 
nitrogen over long periods of time. 

Monoclonal antibodies can be prepared from the above-described 
hybridomas by, for example, a routine procedure of culturing the 

25 hybridomas and obtaining antibodies from the culture supernatants . 
Alternatively, monoclonal antibodies can be prepared by injecting 
the hybridomas into a compatible mammal; growing these hybridomas 
in the mammal; and obtaining antibodies from the mammal's ascites. 
The former method is suitable for preparing highly purified antibodies, 

30 while the latter is suitable for preparing antibodies on a large scale. 

Recombinant antibodies can also be prepared by: cloning an 
antibody gene from a hybridoma; inserting the gene into an appropriate 
vector; introducing the vector into a host; and producing the 
antibodies by using genetic recombination techniques (see, for 

35 example, Vandamme, A. M. et al., Eur. J. Biochem. 192, 767-775, 
(1990) ) . 



47 



Specifically, an mRNA encoding the variable (V) region of 
anti-Mpl antibody is isolated from hybridomas producing the anti-Mpl 
antibodies. For mRNA isolation, total RNAs are first prepared by 
conventional methods such as guanidine ultracentrif ugation methods 
(Chirgwin, J. M. et al., Biochemistry 18, 5294-5299 (1979)), or acid 
guanidinium thiocyanate-phenol-chlorof orm (AGPC) methods 
(Chomczynski, P. et al., Anal. Biochem. 162, 156-159 (1987)), and 
then the target mRNA is prepared using an mRNA Purification Kit 
(Pharmacia) and such. Alternatively, the mRNA can be directly 
prepared using the QuickPrep mRNA Purification Kit (Pharmacia) . 

A cDNA of the antibody V region is synthesized from the resulting 
mRNA using reverse transcriptase. cDNA synthesis is carried out 
using the AMV Reverse Transcriptase First-strand cDNA Synthesis Kit 
(Seikagaku Co. ) , or such. Alternatively, cDNA can be synthesized and 
amplified by the 5' -RACE method (Frohman, M. A. et al., Proc. Natl. 
Acad. Sci. USA 85, 8998-9002 (1988); Belyavsky, A. et al., Nucleic 
Acids Res. 17, 2919-2932 (1989)) using the 5'-Ampli FINDER RACE Kit 
(Clontech) and PCR. 

Target DNA fragments are purified from the obtained PCR products 
and then ligated with vector DNAs to prepare recombinant vectors. 
The vectors are introduced into E. coli and such, and colonies are 
selected for preparing the recombinant vector of interest. The 
target DNA nucleotide sequence is then confirmed by conventional 
methods such as the dideoxynucleot ide chain termination method. 

Once a DNA encoding the V region of target anti-Mpl antibody 
is obtained, the DNA is inserted into an expression vector which 
comprises a DNA encoding the constant region (C region) of a desired 
antibody. 

The method for producing anti-Mpl antibodies to be used in the 
present invention typically comprises the steps of: inserting an 
antibody gene into an expression vector, so that the gene is expressed 
under the regulation of expression regulatory regions, such as 
enhancer and promotor ; and transforming host cells with the resulting 
vectors to express antibodies. 

For expressing the antibody gene, polynucleotides encoding H 
chain and L chain, respectively, are inserted into separate expression 



48 



vectors and co-transf ected into a host cell. Alternatively, 
polynucleotides encoding both H chain and L chain are inserted into 
a single expression vector and transfected into a host cell (see WO 
94/11523) . 

5 The term "agonistic activity" refers to an activity to induce 

changes in some, biological activities through signal transduction 
.into cells and such, due to the binding of an antibody to a receptor 
antigen. The biological activities include, for example, 
proliferation-promoting activities, proliferation activities, 
10 viability activities, differentiation-inducing activities, 
differentiation activities, transcriptional activities, membrane 
transport activities, binding activities, proteolytic activities, 
phosphorylation/dephosphorylation activities, oxidation/ reduction 
activities, transfer activities, nucleolytic activities, 
15 dehydration activities, cell death-inducing activities, and 
apoptosis-inducing activities, but is not limited thereto. 

The term "agonistic activity against Mpl" typically refers to 
the activity of promoting the differentiation of megakaryocytes or 
their parental hemopoietic stem cells into platelets, or the activity 
20 of stimulating platelet proliferation. 

Agonistic activity can be assayed by methods known to those 
skilled in the art. The agonistic activity may be determined using 
the original activity or a different activity as an indicator. 

For example, agonistic activity can be determined by a method 
25 using cell growth as an indicator as described in Examples. More 
specifically, an ant ibody whose agonistic activity is to be determined 
is added to cells which proliferate in an agonist-dependent manner, 
followed by incubation of the cells. Then, a reagent such as WST-8 
which shows a coloring reaction at specific wavelengths depending 
30 on the viable cell count, is added to the culture and absorbance is 
measured. The agonistic activity can be determined using the 
measured absorbance as an indicator. 

Cells that proliferate in an agonist-dependent manner can also 
be prepared by methods known to those skilled in the art . For example, 
35 when the antigen is a receptor capable of transducing cell growth 
signals, cells expressing the receptor may be used. Alternatively, 



49 



when the antigen is a receptor that cannot transduce signals, a 
chimeric receptor consisting of the intracellular domain of a receptor 
that transduces cell growth signals and the extracellular domain of 
a receptor that does not transduce cell growth signals can be prepared 
for cellular expression. Receptors that transduce cell growth 
signals include, for example, G-CSF receptors, mpl, neu, GM-CSF 
receptors, EPO receptors, c-kit, and FLT-3. Cells that can be used 
to express a receptor include, for example, BaF3, NFS 60, FDCP-1, 
FDCP-2, CTLL-2, DA-1, and KT-3. 

There is no limitation as to the type of detection indicators 
to be used for determining agonistic activity, as long as the indicator 
can monitor quantitative and/or qualitative changes. For example, 
it is possible to use cell-free assay indicators, cell-based assay 
indicators, tissue-based assay indicators, and in vivo assay 
indicators. Indicators that can be used in cell-free assays include 
enzymatic reactions, quantitative and/or qualitative changes in 
proteins, DNAs, or RNAs. Such enzymatic reactions include, for 
example, amino acid transfers, sugar transfers, dehydrations, 
dehydrogenations, and substrate cleavages. Alternatively, protein 
phosphorylations, dephosphorylations, dimerizations, 

multimerizations, hydrolyses, dissociations and such; DNA or RNA 
amplifications, cleavages, and extensions can be used as the indicator 
in cell-free assays. For example, protein phosphorylations 
downstream of a signal transduction pathway may be used as a detection 
indicator. Alterations in cell phenotype, for example, quantitative 
and/or qualitative alterations in products, alterations in growth 
activity, alterations in cell number, morphological alterations, or 
alterations in cellular properties, can be used as the indicator in 
cell-based assays. The products include, for example, secretory 
proteins, surface antigens, intracellular proteins, andmRNAs. The 
morphological alterations include, for example, alterations in 
dendrite formation and/or dendrite number, alteration in cell 
flatness, alteration in cell elongation/axial ratio, alterations in 
cell size, alterations in intracellular structure, 
heterogeneity/homogeneity of cell populations, and alterations in 
cell density. Such morphological alterations can be observed under 



50 



a microscope. Cellular properties to be used as the indicator include 
anchor dependency, cytokine-dependent response, hormone dependency, 
drug resistance, cell motility, cell migration activity, pulsatory 
activity, and alteration in intracellular substances . Cell motility 
5 includes cell infiltration activity and cell migration activity . The 
alterations in intracellular substances include, for example, 
alterations in enzyme activity, mRNA levels, levels of intracellular 
signaling molecules such as Ca 2+ and cAMP, and intracellular protein 
levels. When a cell membrane receptor is used, alterations in the 

10 cell proliferating activity' induced by receptor stimulation can be 
used as the indicator. The indicators to be used in tissue-based 
assays include functional alterations adequate for the subject tissue. 
In in vivo assays, alterations in tissue weight, alterations in the 
blood system (for example, alterations in blood cell counts, protein 

15 contents, or enzyme activities), alterations in electrolyte levels, 
and alterations in the circulating system (for example, alterations 
in blood pressure or heart rate) . 

The methods for measuring such detection indices are not 
particularly limited. For example, absorbance, luminescence, color 

20 development, fluorescence, radioactivity, fluorescence polarization, 
surface plasmon resonance signal, time-resolved fluorescence, mass, 
absorption spectrum, light scattering, and fluorescence resonance 
energy transfer may be used. These measurement methods are known to 
those skilled in the art and may be selected appropriately depending 

25 on the purpose. For example, absorption spectra can be obtained by 
using a conventional photometer, plate reader, or such; luminescence 
can be measured with a luminometer or such; and fluorescence can be 
measured with a fluorometer or such. Mass can be determined with a 
mass spectrometer. Radioactivity can be determined with a device 

30 such as a gamma counter depending on the type of radiation. 
Fluorescence polarization can be measured with BEACON (TaKaRa). 
Surface plasmon resonance signals can be obtained with BIACORE. 
Time-resolved fluorescence, fluorescence resonance energy transfer, 
or such can be measured with ARVO or such. Furthermore, a flow 

35 cytometer can also be used for measuring. It is possible to use one 
of the above methods to measure two or more different types of 



detection indices. A greater number of detection indices may also 
be examined by using two or more measurement methods simultaneously 
and/or consecutively. For example, fluorescence and fluorescence 
resonance energy transfer can be measured at the same time with a 
f luoronteter . 

The present invention also provides pharmaceutical 
compositions comprising antibodies of this invention. The 
pharmaceutical compositions comprising antibodies of the present 
invention are useful for treating and/or preventing thrombocytopenia 
and such. Time required for the platelet count to recover to the 
normal level can be shortened by administering an antibody of the 
present invention after donation of platelet components. The amount 
of platelet components at the time of blood collection can be increased 
by pre-administering an antibody of the present invention. 

When used as pharmaceutical compositions, the antibodies of the 
present invention can be formulated by methods known to those skilled 
in the art. For example, the antibodies can be administered 
parenterally by injection of a sterile solution or suspension in water 
or other pharmaceutically acceptable solvents. For example, the 
antibodies can be formulated by appropriately combining with 
pharmaceutically-acceptable carriers or solvents, specifically, 
sterile water or physiological saline, vegetable oils, emulsifiers, 
suspending agents, surfactants, stabilizers, flavoring agents, 
excipients, vehicles, preservatives, binding agents, and such, and 
mixing at a unit dosage and form required by accepted pharmaceutical 
implementations. In such formulations, the amount of the thus 
obtained active ingredient should be within the required range. 

A sterile composition to be injected can be formulated using 
a vehicle such as distilled water used for injection, according to 
standard protocols. 

Aqueous solutions used for injections include, for example, 
physiological saline and isotonic solutions comprising glucose or 
other adjunctive agents such as D-sorbitol, D-mannose, D-mannitol, 
and sodium chloride. They may also be combined with an appropriate 
solubilizing agent such as alcohol, specifically, ethanol, 
polyalcohol such as propylene glycol or polyethylene glycol, or 



52 



non-ionic detergent such as polysorbate 80 or HCO-50, as necessary. 

Oil solutions include sesame oils and soybean oils, and can be 
combined with solubilizing agents such as benzyl benzoate or benzyl 
alcohol. Injection solutions may also be formulated with buffers, 
5 for example, phosphate buffers or sodium acetate buffers; analgesics, 
for example, procaine hydrochloride; stabilizers, for example, benzyl 
alcohol or phenol; or anti-oxidants . The prepared injections are 
typically aliquoted into appropriate ampules. 

The administration is preferably carried out parenterally , 

10 specifically, by injection, intranasal administration, 
intrapulmonary administration, percutaneous administration, or such 
Injections include, for example, intravenous injections, 
intramuscular injections, intraperitoneal injections, and 
subcutaneous injections. The injection solutions can be also 

15 administered systemically or locally. 

The administration methods can be selected properly according 
to the patient's age, condition, and such. The applied dose of a 
pharmaceutical composition comprising an antibody or polynucleotide 
encoding the antibody may be, for example, in the range of 0.0001 

20 to 1,000 mg/kg body weight. Alternatively, the dosage may be, for 
example, in the range of 0 . 001 to 100, 000 mg/kg body weight . However, 
the dosage is not restricted to the values described above. The dosage 
and administration methods depend on the patient's weight, age, and 
condition, and are appropriately selected by those skilled in the 

25 art. 

Furthermore, the present invention relates to methods for 
inducing signals in Mpl-expressing cells by using the antibodies of 
the present invention. More specifically, the present invention 
relates methods for inducing signals in Mpl-expressing cells, in which 
30 the methods comprise the step of contacting the cells with the 
antibodies of the present invention. 

All patents, published patent applications, and publications 
cited herein are incorporated by reference in their entirety. 

35 Examples 

The present invention is specifically illustrated below with 



53 



reference to Examples, but it is not to be construed as being limited 
thereto . 

[Example 1] Preparation of anti-human Mpl antibodies 
1.1 Establishment of Mpl-expressing BaF3 cell lines 

BaF3 cell lines expressing the full-length Mpl gene were 
established to obtain cell lines that proliferate in a TPO-dependent 
manner . 

A full-length human Mpl cDNA (Palacios, R. et al., cell, 41, 
727-734 (1985)) (GenBank accession NO. NM_005373) was amplified by 
PCR. The cDNA was cloned into a pC0S2 expression vector to construct 
pCOS2-hMplf ull . The expression vector pC0S2 was constructed by 
removing the DHFR gene expression region from pCHOI (Hirata, Y. et 
al., FEBS Letter, 356, 244-248 (1994)), where the expression region 
of the neomycin resistance gene HEF-VH-g^l (Sato, K. et al., Mol 
Immunol., 31, 371-381 (1994)) is inserted. 

The cynomolgus monkey Mpl cDNA (SEQ ID NO: 164) was cloned from 
total RNA extracted from the bone marrow cells of cynomolgus monkey, 
using a SMART RACE cDNA Amplification Kit (Clontech) . The resulting 
cynomolgus monkey cDNA was inserted into pC0S2 to construct 
pC0S2-monkeyMplf ull . 

Then, the full-length mouse Mpl cDNA (GenBank accession NO. 
NM_010823) was amplified by PCR, and inserted into pC0S2 to construct 
pC0S2-mouseMplf ull. 

Each vector (20 \xg) prepared as described above was mixed with 
BaF3 cells (lx 10 7 cells/mL) suspended in PBS in Gene Pulser cuvettes. 
This mixture was then pulsed at 0.33 kV and 950 fiFD using a Gene Pulser 
II (Bio-Rad) . The BaF3 cells introduced with the above DNAs by 
electroporation were added to RPMI 1640 medium (Invitrogen) 
containing 1 ng/mL mouse interleukin 3 (hereinafter abbreviated as 
mIL-3; Peprotech) , 500 ug/mL Geneticin (Invitrogen), and 10% FBS 
(Invitrogen), and selected to establish a human Mpl-expressing BaF3 
cell line (hereinafter abbreviated as "BaF3-human Mpl"), monkey 
Mpl-expressing BaF3 cell line (hereinafter abbreviated as BaF3-monkey 
Mpl) , and mouse Mpl-expressing BaF3 cell line (hereinafter 
abbreviated as "BaF3-mouse Mpl") . Following selection, these cells 



54 



were cultured and maintained in RPMI 1640 containing 1 ng/mL rhTPO 
(R&D) and 10% FBS. 

1.2 Establishment of Mpl-expressing CHO cell lines 

5 CHO cell lines expressing the full-length Mpl gene were 

established to obtain cell lines to be used for assessing binding 
activity by flow cytometry. 

First, the DHFR gene expression site from pCHOI was inserted 
into pCXN2 (Niwa, H. et al., Gene, 108, 193-199 (1991) ) at the Hindlll 

10 site to prepare a pCXND3expression vector. The respective Mpl genes 
were amplified by PCR using pC0S2-hMpl f ull, pC0S2-monkeyMplf ull, and 
pCOS2-mouseMplfull as templates, and primers with a His-tag sequence . 
The PCR products were cloned into pCXND3 to construct pCXND3-hMpl-His , 
pCXND3-monkey Mpl-His, and pCXND3-mouse Mpl-His, respectively. 

15 Vectors thus prepared (25 ug each) were mixed with a PBS 

suspension of CHO-DG44 cells (lx 10 1 cells/mL) in Gene Pulser cuvettes . 
The mixture was then pulsed at 1.5 kV and 25 uTD using Gene Pulser 
II (Bio-Rad) . The CHO cells introduced with these DNAs by 
electroporation were added to CHO-S-SFMII medium (Invitrogen) 

20 containing 500 ug/mL Geneticin and lx HT (Invitrogen). A human 
Mpl-expressing CHO cell line (hereinafter abbreviated as "CHO-human 
Mpl"), monkey Mpl-expressing CHO cell line (hereinafter abbreviated 
as "CHO-monkey Mpl"), and mouse Mpl-expressing CHO cell line 
(hereinafter abbreviated as "CHO-mouse Mpl" ) were established through 

25 selection. 

1.3 Preparation of soluble human Mpl protein 

To prepare soluble human Mpl protein, an expression system using 
insect Sf9 cells for production and secretion of the protein was 
30 constructed as described below. 

A DNA construct encoding the extracellular region of human Mpl 
(Gin 26 to Trp 4 91) with a downstream FLAG tag was prepared. The 
construct was inserted into a pBACSurf-1 Transfer Plasmid (Novagen) 
between the PstI and Smal sites to prepare pBACSurf 1-hMpl-FLAG . Then, 
35 Sf9 cells were transformed with 4 |ig of pBACSurf 1-hMpl- FLAG using 
the Bac-N-Blue Transfection Kit (Invitrogen) . The culture 



55 



supernatant was collected after a three-day incubation. Recombinant 
virus was isolated by plaque assays. The prepared virus stock was 
used to infect Sf9 cells, and the culture supernatant was collected. 

Soluble human Mpl protein was purified from the obtained culture 
5 supernatant as described below. The culture supernatant was loaded 
onto a Q Sepharose Fast Flow (Amersham Biosciences) for adsorption, 
and the adsorbed protein was then eluted with 50 mM Na-phosphate buffer 
(pH7.2) containing 0.01% (v/v) Tween 20 and 500 mM NaCl. After the 
eluates were loaded onto a FLAG M2-Agarose (Sigma-Aldrich) for 

10 adsorption, the protein adsorbed was eluted with 100 mM glycine-HCl 
buffer (pH3.5) containing 0.01% (v/v) Tween 20. Immediately after 
elution, the fraction obtained was neutralized with 1 M Tris-HCl 
Buffer (pH8.0) and the buffer was exchanged with PBS(-) and 0.01% 
(v/v) Tween 20 using PD-10 columns (Amersham Biosciences) . The 

15 purified soluble Mpl protein was referred to as "shMpl-FLAG" . 

1.4 Preparation of human Mpl-IgG Fc fusion protein 

Human fusion protein Mpl-IgG Fc gene was prepared according to 
the method by Bennett et al. (Bennett, B. D. et al., J. Biol. Chem. 

20 266, 23060-23067 (1991) ) . A nucleotide sequence encoding the 
extracellular region of human Mpl (Gin 26 to Trp 491) was linked to 
a nucleotide sequence encoding the Fc region of human IgG-yl (a region 
downstream of Asp 216) . A BstEII sequence (amino acids: Val-Thr) was 
attached to the junction as a fusion linker between these two regions. 

25 A 19-amino acid signal peptide derived form human IgG H chain variable 
region was used as the signal sequence. The resulting human fusion 
protein Mpl-IgG Fc gene was cloned into pCXND3 to construct 
pCXND3-hMpl-Fc. 

The vector thus prepared (25 ug) was mixed with a PBS suspension 

30 of CHO-DG44 cells (lx 10 7 cells/mL) in Gene Pulser cuvettes. The 
mixture was then pulsed at 1.5 kV and 25 uFD using Gene Pulser II 
(Bio-Rad) . The CHO cells introduced with the DNA by electroporation 
were added to CHO-S-SFMII medium containing 500 ug/mL Geneticin and 
lx HT (Invitrogen) . shMPL-Fc-expressing CHO cell line (CHO-hMpl-Fc) 

35 was then established through selection. 

Human Mpl-IgG Fc fusion protein was purified from the culture 



56 



supernatant as described below. 

The culture supernatant was loaded onto a Q Sepharose Fast Flow 
(Amersham Biosciences) for adsorption, and then the adsorbed protein 
were eluted with 50 mM Na-phosphate buffer (pH7.6) containing 0.01% 
(v/v) Tween 20 and 1 M NaCl. After the eluates were loaded onto a 
HiTrap protein G HP column (Amersham Biosciences) for adsorption, 
the adsorbed protein was eluted with 0.1 M glycine-HCl buffer (pH2.7) 
containing 150 mM NaCl and 0.01% (v/v) Tween 20. Immediately after 
elution, the obtained fraction was neutralized with 1 M Tris-HCl 
Buffer (pHB.0) and the buffer was exchanged with PBS(-) and 0.01% 
(v/v) Tween 20 using PD-10 columns (Amersham Biosciences) . The 
purified soluble Mpl protein was referred to as "hMpl-Fc". 

1.5 Immunization with shMpl-FLAG or BaF3-human Mpl and hybridoma 
selection 

MRL/MpJUmmCrj-lpr/lpr mice (hereinafter abbreviated as 
"MRL/lpr mice"; purchased from Charles River, Japan) were immunized; 
the primary immunization was carried out at eight weeks of age. For 
every single mouse, an emulsion containing 100 ug of shMPL-FLAG 
combined with Freund' s complete adjuvant (H37 Ra; Beckton Dickinson) , 
was administered subcutaneously as the primary injection. As a 
booster injection, an emulsion containing shMPL-FLAG (50 fig per mouse) 
combined with Freund' s incomplete adjuvant (Beckton Dickinson) was 
administered subcutaneously. Three mice which have been immunized 
six times in total were subjected to a final injection of shMPL-FLAG 
(50 |ag per mouse) through the caudal vein. Cell fusion was achieved 
by mixing the mouse myeloma P3-X63Ag8Ul cells (P3U1; purchased from 
ATCC) and mouse splenocytes using polyethylene glycol 1500 (Roche 
Diagnostics) . Hybridoma selection in HAT medium began the following 
day and culture supernatants were obtained. Screening was carried 
out by ELISA, using immunoplates immobilized with shMpl-FLAG or 
hMpl-Fc and the assayed cell growth activity of BaF3-human Mpl as 
an index. In addition, Balb/C mice were immunized eleven times in 
total by administering BaF3-human Mpl (l.Ox 10 7 cells per mouse) 
intraperitoneally over a period of one week to five months. 
Hybridomas were similarly prepared by cell fusion, and screened using 



57 



the assayed cell growth activity of BaF3-human Mpl as an index. 
Positive clones were isolated as single clones by limiting dilution 
and then cultured in a large scale. The culture supernatants were 
collected. 

5 

1.6 Analyses of anti-human Mpl antibodies 

Antibody concentrations were determined by carrying out a mouse 
IgG sandwich ELISA using goat anti-mouse IgG (gamma) (ZYMED) and 
alkaline phosphatase-goat anti-mouse IgG (gamma) (ZYMED), generating 
10 a calibration curve by GraphPad Prism (GraphPad Software; USA) , and 
calculating the antibody concentrations from the calibration curve. 
Commercially available antibodies of the same isotype were used as 
standards . 

Antibody isotypes were determined by antigen-dependent ELISA 
15 using isotype-specif ic secondary antibodies. hMpl-Fc was diluted to 
1 Ug/mL with a coating buffer (0.1 mM NaHCOj, pH9.6) containing 0 . 02% 
(w/v) NaNa, and then added to ELISA plates . The plates were incubated 
overnight at 4°C for coating. The plates were blocked with a diluent 
buffer (50 mM Tris-HCl (pH8.1) containing 1 mM MgCl 2 , 150 mM NaCl, 
20 0.05% (v/v) Tween 20, 0.02% (w/v) NaN 3 , 1% (w/v) BSA) . After the 
addition of hybridoma culture supernatants, the plates were allowed 
to stand at room temperature for 1 hr. After washing with a rinse 
buffer (0.05% (v/v) Tween 20 in PBS), alkaline phosphatase-labeled 
isotype-specif ic secondary antibodies were added to the plates. Then, 
25 the plates were allowed to stand at room temperature for 1 hr. Color 
development was carried out using SIGMA104 ( Sigma-Aldrich) diluted 
to 1 mg/mL with a substrate buffer (50 mM NaHC0 3 , pH9.8) containing 
10 mM MgCl2/ and absorbance was measured at 405 nm using Benchmark 
Plus (Bio-Rad) . 

30 The binding activities of an antibody to shMpl-FLAG and hMPL-Fc 

were determined by ELISA. ELISA plates were coated with 1 Ug/mL of 
purified shMpl-FLAG or hMPL-Fc, and blocked with a diluent buffer. 
Hybridoma culture supernatants were added to the plates, and the 
plates were allowed to stand at room temperature for 1 hr. Then, 

35 alkaline phosphatase-labeled anti-mouse IgG antibodies (Zymed) were 
added to the plates. Color development was similarly carried out 



58 



using the above method. Following a one-hour coloring reaction at 
room temperature, absorbance was measured at 405 nm and EC50 values 
were computed using GraphPad Prism. 

CHO-human Mpl cells and CHO-monkey Mpl cells were harvested, 
and suspended in FACS Buffer (1% FBS/ PBS) to a final concentration 
of lx 10 6 cells /mL. The suspensions were aliquoted into Multiscreen 
(Millipore) at 100 ul/well, and the culture supernatants were removed 
by centrifugation. Culture supernatants diluted to 5 fig/mL were 
added to the plates and incubated on ice for 30 min. The cells were 
washed once with FACS buffer, and incubated on ice for 30 min following 
the addition of an FITC-labeled anti-mouse IgG antibody (Beckman 
Coulter) . After incubation, the mixture was centrifuged at 500 rpm 
for 1 min. The supernatants were removed, and then the cells were 
suspended in 400 UL of FACS buffer. The samples were analyzed by flow 
cytometry using EPICS ELITE ESP (Beckman Coulter) . An analysis gate 
was set on the forward and side scatters of a histogram to include 
viable cell populations. 

Agonistic activities of an antibody were evaluated using 
BaF3-human Mpl and BaF3-monkey Mpl which proliferate in a 
TPO-dependent manner. Cells of each cell line were suspended at 4x 
10 5 cells/ml in RPMI 1640/10% FBS (Invitrogen) , and each suspension 
was aliquoted into a 96-well plate at 60^1 /well. A 40-\ih aliquot of 
rhTPO (R&D) and hybridoma culture supernatants prepared at various 
concentrations was added into each well. The plates were then 
incubated at 37°C under 5% C0 2 for 24 hr. A 10-uL aliquot of the Cell 
Count Reagent SF (Nacalai Tesque) was added into each well. After 
incubation for 2 hr, absorbance was measured at 4 50 nm (and at 655 
nm as a control) using a Benchmark Plus. EC50 values were calculated 
using GraphPad Prism. 

The above analysis yielded a total of 163 clones of mouse 
monoclonal antibodies that bind to human Mpl. 

Among the anti-human Mpl antibodies to be described, TR136 was 
established from mice immunized with BaF3-human Mpl and the others 
were established from mice immunized with shMpl-Flag. 

1.7 Purification of anti-human Mpl antibodies 



59 



Anti-human Mpl antibodies were purified from hybridoma culture 
supernatants as described below. 

After the culture supernatants were loaded onto HiTrap protein 
G HP columns (Amersham Biosciences) for adsorption, the antibodies 
5 were eluted with 0.1 M glycine-HCl <pH2. 7) Buffer. Immediately after 
elution, the fractions were neutralized with 1 M Tris-HCl Buffer 
(pH9.0), and dialyzed against PBS to replace the buffer for one day. 

1.8 Determination of epitopes for the anti-human Mpl antibody VB22B 

10 Since the anti-human Mpl antibody VB22B can be used for Western 

blotting, a GST-fusion protein containing a partial sequence of human 
Mpl was constructed for VB22B epitope analysis. MG1 (Gln26 to Trp491 ) 
and MG2 (Gln26 to Leu274) regions were each amplified by PCR, and 
cloned into pGEX-4T~3 (Amersham Biosciences) to be expressed as GST 

15 fusion proteins. The resulting plasmid DNAs were transformed into 
DHSa to give transf ormants . A final concentration of 1 mM IPTG was 
added to the transf ormants in their logarithmic growth phase to induce 
the expression of GST fusion proteins. The bacterial cells were 
harvested after two hours of incubation. The cells were lysed by 

20 sonication. The lysates were centrifuged in XL-80 Ultracentrif uge 
(Beckman, Rotor 70.1Ti) at 35,000 rpm for 30 min. The culture 
supernatants were removed, and then the fusion proteins were purified 
using GST Purification Modules (Amersham Biosciences) . The samples 
were separated by 1 0%-SDS-PAGE, and then transferred onto a PVDF 

25 membrane. The membrane was analyzed by the murine antibody VB22B in 
Western Btotting. VB22B was found to recognize both MG-1 and MG-2, 
indicating that the VB22B epitope is located in the (Gln26 to Leu274) 
region . 

Then, GST fusion proteins containing the respective regions of 
30 human Mpl: MG3 (Gln26 to Alal89) , MG4 (Gln26 to Prol06) , MG5 (Gln26 
to Glu259) , and MG6 (Gln2S to Gly245) were prepared and analyzed by 
Western blotting using the same procedure described above. VB22B was 
found to recognize MG5 and MG6, but not MG3 and MG4 . This suggests 
that the VB22B epitope is located within the (Alal89 to Gly245) region. 
35 In addition, GST was fused with MG7 (Gln26 to Ala231) and MG8 (Gln26 
to Pro217) to prepare GST fusion proteins. VB22B recognized MG7 but 



60 



not MG8, suggesting that the VB22B epitope is located in the (Gln217 
to Ala231) region. Furthermore, GST fusion protein containing MG10 
(Gln213 to Ala231) was recognized by VB22B, suggesting that the VB22B 
epitope is located within the limited region of 19 amino acids between 
5 Gln213 and Ala231. 

1.9 Kinetic analyses of the antigen-antibody reaction for anti-human 
Mpl antibody VB22B 

Since the anti-human Mpl antibody VB22B binds to soluble 

10 recombinant Mpl, kinetic analyses of the antigen-antibody reaction 
between VB22B IgG and human Mpl-IgG Fc fusion protein were carried 
out as described in Example 1.4. The Sensor Chip CM 5 (Biacore) was 
placed in Biacore 2000 (Biacore) , and human Mpl-IgG Fc fusion protein 
was immobilized onto the chip by amine-coupling methods. Then, 1.25 

15 to 20 ug/mL of VB22B IgG solution was prepared using HBS-EP Buffer 
(Biacore), and injected over the chip surface for 2 min to reveal 
the binding region. Then, HBS-EP Buffer was injected over the chip 
surface for 2 min to reveal the dissociation region. VB22B IgG bound 
to the human Mpl-IgG Fc fusion protein on the sensor chip was removed 

20 by injecting 10 mM NaOH over the sensor chip for 15 sec, and the chip 
was recovered. HBS-EP Buffer was used as the running buffer, and the 
flow rate was 20 nL/min. Using the BIAevaluation Version 3.1 
(Biacore) software, the reaction rate constant at each concentration 
was calculated from the sensorgrams. The dissociation constant (KD) 

25 for VB22B IgG was determined to be 1.67 ± 0.713 x 10" 9 M. 

[Example 2] Preparation of single-chain anti-human Mpl antibodies 
Among the prepared anti-human Mpl antibodies, 23 types of 
antibodies, which exhibit higher binding activities and agonistic 
30 activities, were selected to construct expression systems for 
single-chain antibodies using genetic engineering technigues. An 
exemplary method for constructing a single-chain antibody derived 
from the anti-human Mpl antibody VB22B is described below. 

35 2.1 Cloning of the anti-human Mpl antibody variable region 

The variable region was amplified by RT-PCR using total RNA 



61 



extracted from hybridomas producing anti-human Mpl antibodies. 
Total RNA was extracted from lx 10 7 hybridoma cells using the RNeasy 
Plant Mini Kit (QIAGEN) . 

A 5' -terminal fragment of the gene was amplified from 1 ug of 
5 total RNA by the SMART RACE cDNA Amplification Kit (Clontech) , using 
a synthetic oligonucleotide MHC-IgG2b (SEQ ID NO: 166) complementary 
to mouse IgG2b constant region or a synthetic oligonucleotide kappa 
(SEQ ID NO: 167) complementary to mouse k chain constant region. 
Reverse transcription was carried out at 42°C for 1.5 hr . 
10 The composition of the PCR reaction solution (50 (1L in total) 

is shown below. 



lOx Advantage 2 PCR Buffer (Clontech) 




5 


ML 


lOx Universal Primer A Mix (Clontech) 




5 


HL 


dNTPs (dATP, dGTP, dCTP, and dTTP) (Clontech) 


0 


2 


mM 


Advantage 2 Polymerase Mix (Clontech) 




1 


ML 


Reverse transcription product 


2 


5 


ML 


Synthetic oligonucleotide, MHC-IgG2b or kappa 


10 


pmol 



The PCR reaction conditions were: 

94°C (initial temperature) for 30 sec; 
15 five cycles of 94°C for 5 sec and 72°C for 3 min; 

five cycles of 94 °C for 5 sec, 70°C for 10 sec, and 72°C for 3 min; 

25 cycles of 94°C for 5 sec, 68°C for 10 sec, and 72°C for 3 min; 

and final extension was at 72°C for 7 min. 

The PCR products were purified from agarose gel using the 
20 QIAquick Gel Extraction Kit (QIAGEN), and cloned into a pGEM-T Easy 

Vector (Promega) . The nucleotide sequence was then determined using 

the ABI 3700 DNA Analyzer (Perkin Elmer). 

The nucleotide sequence of cloned VB22B H chain variable region 

(hereinafter abbreviated as "VB22B-VH" ) is shown in SEQ ID NO: 117, 
25 and its amino acid sequence is shown in SEQ ID NO: 118. The nucleotide 

sequence of the L chain variable region (hereinafter abbreviated as 

"VB22B-VL") is shown in SEQ ID NO: 119, and its amino acid sequence 

is shown in SEQ ID NO: 120. 



62 



2.2 Preparation of expression vectors for anti-human Mpl diabodies 
The gene encoding VB22B single-chain Fv (hereinafter 
abbreviated as "VB22B diabody") containing a five-amino acid linker 
sequence was constructed, by linking a nucleotide sequence encoding 
5 a (Gly«Ser)i linker to the VB22B-VH-encoding gene at its 3' end and 
to the VB22B-VL-encoding gene at its 5' end; both of which have been 
amplified by PCR. 

The VB22B-VH forward primer, 70-115HF, ( SEQ ID NO: 168) was 
designed to contain an EcoRI site. The VB22B-VH reverse primer, 

10 33-115HR, (SEQ. ID NO: 169) was designed to hybridize to a DNA encoding 
the C terminus of VB22B-VH, and to have a nucleotide sequence encoding 
the (Gly^Ser) i linker and a nucleotide sequence hybridizing to the 
DNA encoding the N terminus of VB22B-VL. The VB22B-VL forward primer, 
33-115LF, (SEQ ID NO: 170) was designed to have a nucleotide sequence 

15 encoding the N terminus of VB22B-VL, a nucleotide sequence encoding 
the (Gly 4 Ser)i linker, and a nucleotide sequence encoding the C 
terminus of VB22B-VH. The VB22B-VL reverse primer, 33-115LR, (SEQ 
ID NO: 171) was designed to hybridize to a DNA encoding the C terminus 
of VB22B-VL and to have a nucleotide sequence encoding a FLAG tag 

20 (Asp Tyr Lys Asp Asp Asp Asp Lys/SEQ ID NO: 172) and a NotI site. 

In the first round of PCR, two PCR products: one containing 
VB22B-VH and a linker sequence, and the other containing VB22B-VL 
and the identical linker sequence, were synthesized by the procedure 
described below. 

25 The composition of the PCR reaction solution (50 uL in total) 

is shown below. 



lOx PCR Buffer (TaKaRa) 


5 UL 


dNTPs (dATP, dGTP, dCTP, and dTTP) (TaKaRa) 


0 . 4 mM 


DNA polymerase TaKaRa Ex Taq (TaKaRa) 


2 . 5 units 


pGEM-T Easy vector comprising VB22B-VH or 
VB22B-VL gene 


10 ng 


Synthetic oligonucleotides, 70-115HF and 
33-115HR, or 33-115LF and 33-115LR 


10 pmol 


The PCR reaction conditions were: 



63 



94°C (initial temperature) for 30 sec- 
five cycles of: 94°C for 15 sec and 72°C for 2 min; 
five cycles of 94 °C for 15 sec and 70°C for 2 min; 
28 cycles of 94 °C for 15 sec and 68°C for 2 min; 
5 and final extension was at 72°C for 5 min. 

After the PCR products of about 400 bp were purified from agarose 
gel using the QIAquick Gel Extraction Kit (QIAGEN), the second-round 
PCR was carried out using aliquots of the respective PCR products 
according to the protocol described below. 
10 The composition of the PCR reaction solution (50 uL in total) 

is shown below. 



lOx PCR Buffer (TaKaRa) 






5 UL 


dNTPs (dATP, dGTP, dCTP, 


and dTTP) (TaKaRa) 


0 


4 mM 


DNA polymerase TaKaRa Ex 


Taq (TaKaRa) 


2 . 5 


unit 


First-round PCR products 


(two types) 




1 UL 


Synthetic oligonucleotides, 70-115HFand 33-115LR 


10 


pmol 



The reaction conditions were: 

94°C (initial temperature) for 30 sec; 
15 five cycles of 94°C for 15 sec and 72°C for 2 min; 

five cycles of 94 °C for 15 sec and 70°C for 2 min; 

28 cycles of 94 °C for 15 sec and 6B°C for 2 min; 

and final extension was at 72 °C for 5 min. 

The PCR products of about 800 bp were purified from agarose gel 
20 using the QIAquick Gel Extraction Kit (QIAGEN), and then digested 

with EcoRI and NotI (both from TaKaRa) . The resulting DNA fragments 

were purified using the QIAquick PCR Purification Kit (QIAGEN) , and 

then cloned into pCXND3 to prepare pCXND3-VB22B db. 

25 2.3 Preparation of expression vectors for anti-human Mpl antibody 
sc (Fv) 2 

To prepare expression plasmids for the modified antibody 
[sc(Fv)2] comprising two units of H chain variable region and two 
units of L chain variable region derived from VB22B, the 
30 above-described pCXND3-VB22B db was modified by PCR using the 



64 



procedure shown below . The process for constructing the sc ( Fv) 2 gene 
is illustrated in Fig. 1. 

First, PCR method was carried out to amplify (a) the 
VB22B-VH-encoding gene in which a nucleotide sequence encoding a 
5 15-amino acid linker (Gly 4 Ser) 3 was added to its 3' end; and (b) the 
VB22B-VL-encoding gene containing the identical linker nucleotide 
sequence added to its 5' end. The desired construct was prepared by 
linking these amplified genes. Three new primers were designed in 
this construction process'. The VB22B-VH forward primer, VB22B-fpvu, 

10 (primer A; SEQ ID NO: 173) was designed to have an EcoRI site at its 
5' end and to convert Gln22 and Leu23 of VB22B db into a PvuII site. 
The VB22B-VH reverse primer, sc-rL15, (primer B; SEQ ID NO: 174) was 
designed to hybridize to a DNA encoding the C terminus of VB22B-VH, 
and to have a nucleotide sequence encoding the (Gly4Ser) 3 linker, as 

15 well as a nucleotide sequence hybridizing to a DNA encoding the N 
terminus of VB22B-VL. The VB22B-VL forward primer, sc-fL15, (primer 
C; SEQ ID NO: 175) was designed to have a nucleotide sequence encoding 
the N terminus of VB22B-VL, a nucleotide sequence encoding the 
(Gly 4 Ser) 3 linker, and a nucleotide sequence encoding the C terminus 

20 of VB22B-VH. 

In the first-round PCR, two PCR products: one comprising 
VB22B-VH and a linker sequence, and the other comprising VB22B-VL 
and the identical linker sequence, were synthesized by the procedure 
described below. 

25 The composition of the PCR reaction solution (50 UL in total) 

is shown below. 



lOx PCR Buffer (TaKaRa) 


5 \IL 


dNTPs (dATP, dGTP, dCTP, and dTTP) (TaKaRa) 


0.4 mM 


DNA polymerase TaKaRa Ex Taq (TaKaRa) 


2 . 5 units 


pCXND3-VB22B db 


10 ng 


Synthetic oligonucleotides, VB22B-fpvu, sc-rL15 


10 pmol 


or sc-fL15, and 33-115LR (primer D) 





The reaction conditions were: 
94 °C (initial temperature) for 30 sec; 



65 



five cycles of 94°C for 15 sec and 72°C for 2 min; 
five cycles of 94 °C for 15 sec and 70°C for 2 min; 
28 cycles of 94°C for 15 sec and 68°C for 2 min; 
and final extension was at 72°C for 5 rain. 
5 After the PCR products of about 400 bp were purified from agarose 

gel using the QIAquick Gel Extraction Kit (QIAGEN), the second-round 
PCR was carried out using aliquots of the respective PCR products 
according to the protocol described below. 

The composition of the PCR reaction solution (50 UL in total) 
10 is shown below. 



lOx PCR Buffer (TaKaRa) 


5 UL 


dNTPs (dATP, dGTP, dCTP, and dTTP) (TaKaRa) 


0.4 mM 


DNA polymerase TaKaRa Ex Taq (TaKaRa) 


2 . 5 units 


First-round PCR product (two types) 


1 UL 


Synthetic oligonucleotide, 70-115HF and 33-115LR 


10 pmol 


The reaction conditions were: 



94°C (initial temperature) for 30 sec- 
five cycles of 94 °C for 15 sec and 72°C for 2 min; 

15 five cycles of 94°C for 15 sec and 70°C for 2 min; 
28 cycles of 94 °C for 15 sec and 68°C for 2 min; 
and final extension was at 72°C for 5 min. 

The PCR products of about 800 bp were purified from agarose gel 
using the QIAquick Gel Extraction Kit (QIAGEN) , and then digested 

20 with EcoRI and NotI (both from TaKaRa) . The resulting DNA fragments 
were purified using the QIAquick PCR Purification Kit (QIAGEN) , and 
then cloned into pBacPAK9 (Clontech) to construct pBacPAK9-scVB22B . 

A fragment to be inserted into the PvuII site of 
pBacPAK9-scVB22B was prepared. Specifically, the fragment has a 

25 PvuII recognition site at both ends and a nucleotide sequence, in 
which a gene encoding the VB22B-VH N-terminus is linked, via a 
(Gly.iSer)3 linker-encoding nucleotide sequence, to a gene encoding 
the amino acid sequence of an N terminus-deleted VB22B-VH linked to 
VB22B-VL via the (Gly^Ser^ linker. Two primers were newly designed 

30 to prepare the fragment by PCR. The forward primer for the fragment 



66 



of interest, Fv2-f (primer E; SEQ ID NO: 176), was designed to have 
a PvuII site at its 5' end and a VB22B-VH 5' -end sequence. The reverse 
primer for the fragment of interest, Fv2-r (primer F; SEQ ID NO: 177) , 
was designed to hybridize to a DNA encoding the C terminus of VB22B-VL, 
5 and to have a PvuII site, a nucleotide sequence encoding the (Gly«Ser) 3 
linker, and a nucleotide sequence hybridizing to a DNA encoding the 
N terminus of VB22B-VH. PCR was carried out using pBacPAK9-scVB22B 
as a template as described below. 

The composition of the PCR reaction solution (50 UL in total) 
10 is shown below. 



lOx PCR Buffer (TaKaRa) 


5 UL 


dNTPs (dATP, dGTP, dCTP, and dTTP) (TaKaRa) 


0.4 mM 


DNA polymerase TaKaRa Ex Taq (TaKaRa) 


2 . 5 units 


pBacPAK9-scVB22B 


10 ug 


Synthetic oligonucleotide, Fv2-f and Fv2-r 


10 pmol 


The reaction conditions were: 



94 °C (initial temperature) for 30 sec- 
five cycles of 94°C for 15 sec and 72°C for 2 min; 

15 five cycles of 94°C for 15 sec and 70 D C for 2 min; 
28 cycles of 94 °C for 15 sec and 68°C for 2 min; 
and final extension was at 72°C for 5 min. 

The PCR products of about 800 bp were purified from agarose gel 
using the QIAquick Gel Extraction Kit (QIAGEN) , and then cloned into 

20 the pGEM-T Easy Vector (Promega) . After sequencing, the plasmid was 
digested with PvuII (TaKaRa) , and the fragment of interest was 
recovered. The recovered fragment was ligated to pBacPAK9-scVB22B 
pre-digested with PvuII (TaKaRa) to construct pBacPAK9-VB22B sc (Fv) 2 . 
After the resulting vector was digested with EcoRI and NotI (both 

25 from TaKaRa) , the fragment of about 1, 600 bp was purified from agarose 
gel using the QIAquick Gel Extraction Kit (QIAGEN) . The fragment was 
then cloned into a pCXND3 expression vector to construct pCXND3-VB22B 
sc (Fv) 2. 

30 2.4 Expression of single-chain anti-human Mpl antibody in animal cells 



67 



A cell line stably expressing the single-chain antibody was 
prepared from CHO-DG44 cells as described below. Gene transfer was 
achieved by electroporation using a Gene Pulser II (Bio-Rad) . An 
expression vector (25 ug) and 0.75 mL of CHO-DG44 cells suspended 
in PBS (Ix 10 7 cells/mL) were mixed. The resulting mixture was cooled 
on ice for 10 min, transferred into a cuvette, and pulsed at 1.5-kV 
and 25 fiFD. After a ten-minute restoration period at room temperature, 
the electroporated cells were plated in CHO-S-SFMIX medium 
(Invitrogen) containing 500 Hg/mL Geneticin (Invitrogen) . CHO cell 
lines expressing the single-chain antibody were established through 
selection. A cell line stably expressing VB22B sc(Fv)2 and its 
culture supernatants were obtained by this method. 

The transient expression of the single-chain antibody was 
achieved using COS7 cells as described below. An expression vector 
(10 ug) and 0.75 mL of COS7 cells suspended in PBS <lx 10 7 cells/mL) 
were mixed. The resulting mixture was cooled on ice for 10 min, 
transferred into a cuvette, and then pulsed at 1.5-kV and 25 M-FD . 
After a ten-minute restoration period at room temperature, the 
electroporated cells were plated in DMEM/10% FBS medium (Invitrogen) . 
The cells were incubated overnight and then washed with PBS. 
CHO-S-SFMII medium was added and the cells were cultured for about 
three days. The culture supernatants for preparing the VB22B diabody 
were thus prepared. 

2. 5 Quantitation of single-chain anti-human Mpl antibodies in culture 
supernatant s 

The culture supernatant concentration of the single-chain 
anti-human Mpl antibody transiently expressed in COS cells was 
determined using surface plasmon resonance. A sensor chip CM 5 
(Biacore) was placed in Biacore 2000 (Biacore) . ANTI-FLAG® M2 
Monoclonal Antibody ( Sigma-Aldrich) was immobilized onto the chip. 
An appropriate concentration of sample was injected over the chip 
surface at a flow rate of 5 mL/sec, and 50 mM diethylamine was used 
to dissociate the bound antibody. Changes in the mass during sample 
injection were recorded, and the sample concentration was calculated 
from the calibration curve prepared using the mass changes of a 



68 



standard sample. dbl2E10 (see WO 02/33073 and WO 02/33072) was used 
as the diabody standard, and 12E10 sc(Fv)2 which has the same gene 
structure as that of sc(Fv)2 was used as the sc(Fv)2 standard. 

5 2.6 Purification of anti-human Mpl diabodies and single-chain 
antibodies 

The culture supernatants of VB22B diabody-expressing COS7 cells 
or CHO cells was loaded onto an Anti-Flag M2 Affinity Gel 
( Sigma-Aldrich) column equilibrated with a 50 mM Tris-HCl buffer 
10 (pH7.4) containing 150 mM. NaCl and 0.05% Tween 20. The absorbed 
antibodies were eluted with 100 mM glycine-HCl (pH3.5). The 
fractions eluted were immediately neutralized with 1 M Tris-HCl 
(pH8.0), and loaded onto a HiLoad 26/60 Superdex 200 pg (Amersham 
Biosciences) column for gel filtration chromatography. PBS/0.01% 

15 Tween 20 was used in the gel filtration chromatography. 

VB22B sc(Fv)2 was purified from the culture supernatants of 
VB22B sc (Fv) 2-expressing COS7 cells or CHO cells under the same 
conditions used for purifying the diabodies. A large-scale 
preparation of VB22B sc(Fv)2 was prepared by loading the CHO cell 

20 culture supernatants onto a Macro-Prep Ceramic Hydroxyapatite Type 
I (Bio-Rad) column equilibrated with a 20 mM phosphate buffer (pH6.8), 
and eluting the VB22B sc(Fv)2 in a stepwise manner with 250 mM 
phosphate buffer (pH6.8). The eluted fraction was concentrated on 
an ultrafilter, and then fractionated by gel filtration 

25 chromatography using a HiLoad 26/60 Superdex 200 pg (Amersham 
Biosciences) column, and a fraction corresponding to the molecular 
weight range of about 40 kD to 70 kD was obtained. The fraction was 
loaded onto an Anti-Flag M2 Affinity Gel column equilibrated with 
a 50 mM Tris-HCl buffer (pH7.4) containing 150 mM NaCl and 0.05% Tween 

30 20. The absorbed antibody was eluted with 100 mM glycine-HCl (pH3.5). 
The eluted fraction was immediately neutralized with 1 M Tris-HCl 
(pH8.0), and loaded onto a HiLoad 26/60 Superdex 200 pg (Amersham 
Biosciences) column for gel filtration chromatography . 20 mM acetate 
buffer (pH6.0) containing 150 mM NaCl and 0.01% Tween 80 was used 

35 in the gel filtration chromatography. In each purification step, the 
presence of the diabody and sc(Fv)2 in the samples was confirmed by 



69 



SDS-PAGE and Western blotting using an anti-Flag antibody 
(Sigma-Aldrich) . Specifically, obtained fractions corresponding to 
each peak were subjected to the electrophoresis according to the 
method described by Laemli, and then stained using Coomassie Brilliant 
5 Blue. As a result, single band was detected apparently at about 29 
kDa for the diabody; while single band was detected apparently at 
about 55 kDa for sc(Fv)2. 

2.7 Binding activity analyses of single-chain anti-human Mpl 

10 antibodies by flow cytometry 

CHO-human Mpl, CHO-monkey Mpl, and CHO-mouse Mpl cells were 
recovered and suspended in FACS buffer (1% FBS/PBS) to a final 
concentration of Ix 10 6 cells/mL. Cell suspensions were aliquoted 
at 100-^L/well into the Multiscreen-HV Filter Plates (Millipore) . 

15 After centrif ugation, the supernatant was removed. An appropriate 
concentration of diabody or sc<Fv)2 was added into each well and 
incubated on ice for 30 min. The cells were washed once with 200 uL 
of FACS buffer, and incubated on ice for 30 min following the addition 
of 10 U-g/mL ANTI-FLAG® M2 Monoclonal Antibody (Sigma-Aldrich) . The 

20 cells were then washed once with 200 UL of FACS buffer, and a 
lOOx-diluted FITC-labeled anti-mouse IgG antibody (Beckman Coulter) 
was added to the plate. The plate was incubated on ice for 30 min. 
After centrifugation, the supernatant was removed. The cells were 
suspended in 400 uL of FACS Buffer, and then analyzed by flow cytometry 

25 using EPICS ELITE ESP (Beckman Coulter) . An analysis gate was set 
on the forward and side scatters of a histogram to include viable 
cell populations. 

The binding activity of the purified VB22B sc(Fv)2 to various 
Mpl molecules expressed in CHO cells was determined (Fig. 2) . VB22B 
30 sc (Fv) 2 was found to specifically bind to CHO-human Mpl and CHO-monkey 
Mpl but not to the host cell CHO or CHO-mouse Mpl. This binding 
characteristic of VB22B sc(Fv)2 is comparable to those of VB22B IgG, 
indicating that the antibody binding site remains unaltered by 
converting whole IgG to minibody. 



35 



2.8 Analyses of TPO-like agonistic activity for single-chain 



70 



anti-human Mpl antibodies 

TPO-like agonistic activity was assessed using BaF3-human Mpl 
or BaF3-monkey Mpl that proliferate in a TPO-dependent manner. 

Cells from each cell line were washed twice with RPMI 1640/1% 
FBS (fetal bovine serum) ( Invitrogen) , and then suspended in RPMI 
1640/10% FBS to a concentration of 4x 10 5 cells/mL. Cell suspensions 
were aliquoted at 60-uL/well into a 96-well plate. Various 
concentrations of rhTPO (R&D) and COS7 culture supernatants or 
purified samples were prepared, and a 4 0-uL aliquot was added into 
each well. The plates were then incubated at 37 °C under 5% C0 2 for 
24 hr. Immediately after a 10-uL aliquot of WST-8 reagent (Cell Count 
Reagent SF; Nacalai Tesque) was added into each well, absorbance was 
measured at 450 nm (and at 655 ran as a control) using Benchmark Plus. 
After two hours of incubation, absorbance was again measured at 450 
nm (and at 655 nm as a control) . The WST-8 reagent changes colors 
at 450 nm in a color reaction that reflects the viable cell count. 
The TPO-like agonistic activity was assessed using the change in 
absorbance during the two-hour incubation as an index. EC 5 o values 
were computed using GraphPad Prism. 

TPO-like agonistic activity was assayed using the human 
leukemia cell line M-07e (purchased from DSMZ) which proliferates 
TPO-dependently. M-07e cells were washed twice with RPMI 1640/1% FBS, 
and then suspended in RPMI 1640/10% FBS to a concentration of 5x 10 s 
cells/mL. The resulting cell suspension was aliquoted at 50-uL/well 
into a 96-well plate. Various concentrations of rhTPO and COS7 
culture supernatants or purified samples were prepared, and a 50-ui 
aliquot was added into each well. The plates were then incubated at 
37°C under 5% C0 2 for 48 hr. Immediately after a 10-uL aliquot of 
WST-8 reagent (Cell Count Reagent SF; Nacalai Tesque) was added to 
each well, absorbance of was measured at 450 nm (and at 655 nm as 
a control) using a Benchmark Plus. After four hours of incubation, 
absorbance was again measured at 450 nm (and at 655 nm as a control) . 
The TPO-like agonistic activity was assayed using the change in 
absorbance during the four-hour incubation as an index. 

Purified VB22B IgG, VB22B diabody, and VB22B sc(Fv)2 were 
assayed for their TPO-like agonistic activities using BaF3-human Mpl, 



71 



BaF3-monkey Mpl, and M-07e. The results are shown in Figs. 3, 4, and 
5, respectively. The presence of bivalent antigen-binding domains 
in a single antibody molecule is essential for its agonistic activity. 
The distance and angle between two antigen-binding domains can also 
be important factors (see WO 02/33073 and WO 02/33072). Similar 
results were obtained for the newly isolated anti-human Mpl antibodies . 
Specifically, the agonistic activities of VB22B diabody and VB22B 
sc(Fv)2 (EC 50 = 61 pM and 27 pM in BaF3-human Mpl, respectively) were 
higher than that of VB22B IgG (EC 5 o > 30 nM in BaF3-human Mpl), and 
were equivalent to or higher than that of the naturally-occurring 
human TPO ligand (EC 50 = 76 pM in BaF3-human Mpl) . The VB22B diabody 
activity was lower than that of VB22B sc(Fv)2. This suggests that 
the structure of a single-chain antibody is greatly altered by its 
molecular shape and the length of the linker sequence, which in turn 
changes the agonistic activity. Sixteen types of the single-chain 
anti-human Mpl antibodies were obtained, each exhibiting a high 
agonistic activity. The amino acid sequences of the H chain and L 
chain variable regions of the representative antibodies are shown 
in Figs. 6 and 7, respectively. 

2.9 Humanization of single-chain anti-human Mpl antibody 

Antibody sequence data for the humanization of VB22B sc(Fv)2 
were obtained from the Kabat Database 

(ftp://ftp.ebi.ac.uk/pub/databases/kabat/), and homology searches 
were carried out independently for the H chain variable region and 
the L chain variable region. As a result, the H chain variable region 
was found to be highly homologous to DN13 (Smithson S. L. et al., 
Mol Immunol. 36, 113-124 (1999)). The L chain variable region was 
found to be highly homologous to ToP027 (Hougs L. et al., J. Immunol. 
162, 224-237 (1999)). Humanized antibodies were prepared by 
inserting a complementarity-determining region (hereinafter 
abbreviated as "CDR" ) into the framework regions (hereinafter 
abbreviated as "FR") of the above antibodies. The humanized antibody 
sc(Fv)2 was expressed in CHO-DG44 cells, and its agonistic activity 
was assessed using BaF3-human Mpl. The agonistic activity was used 
as an index to generate a humanized VB22B sc(Fv)2 which has agonistic 



72 



activity equivalent to that of murine VB22B sc(Fv)2 by replacing one 
or more amino acids in its framework region. 

Specifically, synthetic oligo-DNAs of approximately 50 
nucleotides in length were designed as to make 20 of these nucleotides 
available for hybridization, and the synthetic oligo-DNAs were 
assembled by PCR to prepare genes that encode the respective variable 
regions. Using the resulting genes, sc(Fv)2 was similarly prepared 
by the method described in Example 2.3. The respective DNAs were 
cloned into a pCXND3expression vector to construct expression vectors 
pCXND3-hVB22B p-z sc(Fv)2, pCXND3-hVB22B g-e sc(Fv)2, pCXND3-hVB22B 
e sc{Fv)2, pCXND3-hVB22B u2-wz4 sc(Fv)2, and pCXND3-hVB22B q-wz5 
sc(Fv)2, to which the humanized VB22B sc(Fv)2 is inserted. The 
nucleotide sequences and the amino acid sequences of the fragments 
in each plasmid are shown below. 

Plasmid name Nucleotide Amino acid 



sequence sequence 



hVB22B p-z sc(Fv)2 


SEQ 


ID NO 


1 


SEQ 


IC 


NO 


2 


hVB22B g-e sc(Fv)2 


SEQ 


ID 


NO: 


253 


SEQ 


ID 


NO: 


254 


hVB22B e sc (Fv) 2 


SEQ 


ID 


NO: 


259 


SEQ 


ID 


NO: 


260 


hVB22B u2-wz4 sc(Fv)2 


SEQ 


ID 


NO: 


286 


SEQ 


ID 


NO: 


287 


hVB22B q-wz5 sc(Fv)2 


SEQ 


ID 


NO: 


292 


SEQ 


ID 


NO: 


293 


Murine VB22B sc(Fv)2 


SEQ 


ID 


NO: 


263 


SEQ 


ID 


NO: 


264 



The plasmids were expressed in CHO-DG44 cells and the culture 
supernatants were recovered by the method described in Example 2.4. 
Since the humanized VB22B sc(Fv)2 does not contain a Flag tag, its 
purification from the culture supernatant was performed using a 
MG10-GST fusion protein. MG10 (Gln213 to Ala231) is one of the 
epitopes recognized by VB22B, as described in Example 1.8. The 
MG10-GST fusion protein was purified using Glutathione Sepharose 4B 
(Amersham Biosciences) according to the supplier's protocol. Then, 
the purified MG10-GST fusion protein was immobilized onto a HiTrap 
NHS-activated HP Column (Amersham Biosciences ) to prepare an affinity 
column, according to the supplier's protocol. The culture 
supernatant of CHO cells expressing the humanized VB22B sc(Fv)2 was 



73 



loaded onto the MG10-GST fusion protein-immobilized column, which 
has been equilibrated with 50 mM Tris-HCl (pH7.4)/150 mM NaCl/0.01% 
Tween 80. The adsorbed humanized VB22B sc(Fv)2 was eluted with 100 
mM glycine-HCl (pH3 . 5 ) /0 . 01% Tween 80. Immediately after elution, 
the eluted fraction was neutralized with 1 M Tris-HCl (pH7 . 4 ) , and 
was further subjected to gel filtration chromatography using a HiLoad 
16/60 Superdex 200 pg (Amersham Biosciences) . 20 mM citrate buffer 
(pH7.5) containing 300 mM NaCl and 0.01% Tween 80 was used in the 
gel filtration chromatography. The TPO-like agonistic activities of 
the purified samples were similarly determined using the method 
described in Example 2.8. The TPO-like agonistic activities of the 
purified murine VB22B sc(Fv)2, hVB22B p-z sc(Fv)2, hVB22B u2-wz4 
sc(Fv)2, hVB22B q-wz5 sc(Fv>2, and humanized hVB22B e sc(Fv}2 and 
hVB22B g-e sc'(Fv) 2 were assessed in BaF3-human Mpl . The results are 
shown in Figs. 19, 20, and 21. The humanized VB22B sc(Fv)2 showed 
comparable agonistic activities, suggesting that the humanization 
has no influence on the activity. 

2.10 Kinetic analyses of the antigen-antibody reaction for anti-human 
Mpl antibodies: VB22B IgG, VB22B sc(Fv)2, and humanized VB22B sc(Fv)2 
Using the soluble recombinant Mpl-binding characteristic of 
anti-human Mpl antibody VB22B, kinetic analyses of the 
antigen-antibody reactions between the MG10 (Gin 213 to Ala 231) -GST 
fusion protein and each of VB22B IgG, VB22B sc(Fv)2, and humanized 
VB22B sc ( Fv) 2 were carried out as described in Example 1.8. The Sensor 
Chip CM5 (Biacore) was placed in Biacore 3000 (Biacore) , and MG10-GST 
fusion protein was immobilized onto the chip by amine-coupling methods 
HBS-EP Buffer (Biacore) was used as the running buffer, and the flow 
rate was 20 uL/min. 5 . 5 to 175 . 0 nM of VB22B IgG solution was prepared 
using HBS-EP Buffer, and injected over each of the chip surfaces for 
2 min to obtain the binding region at the respective concentrations. 
Then, dissociation region for the 2 minutes was measured. VB22B IgG 
bound to the MG10-GST fusion protein on the sensor chip was removed 
by injecting' 20 mM HC1 over the sensor chip for 1 min, and the chip 
was recovered. Similarly, 4.7 to 150.1 nM of VB22B sc(Fv)2, 5.3 to 
168. 9 nM of hVB22B q-wz5 sc(Fv)2, and 4. 9 to 156.8 nM of hVB22B u2-wz4 



74 



sc(Fv)2 were prepared and injected over the chip surfaces onto which 
MG10-GST fusion protein was immobilized, and the measurement was 
carried out. 

All the antibodies used were bivalent antibodies, and thus the 
5 sensorgrams at each concentration were obtained in the presence of 
both monovalent and bivalent bindings. In this context, the reaction 
rate constant was determined as that for the monovalent antibody by 
analysis using the Bivalent analyte model of BIAevaluation ver.3.1 
software (Biacore) . The above analysis was carried out in 

10 triplicates for each antibody. The binding rate constant (ka) , 
dissociation rate constant (kd) , and dissociation constant (KD) were 
determined as those for the monovalent antibody by the procedure 
described above. The constants are indicated below in Table 1. The 
dissociation constants (KD) for VB22B IgG, VB22B sc (Fv) 2, hVB22Bq-wz5 

15 sc(Fv)2, and hVB22B u2-wz4 sc(Fv)2 were determined to be 1.15 x 10" 8 
M, 1.17 x 10" 8 M, 1.36 x 1CT B M, and 1.02 x 1CT B M, respectively, showing 
nearly equivalent binding activities towards the MG10-GST fusion 
protein . 

20 Table 1 Kinetic analyses of the antigen-antibody reaction for 
anti-human Mpl antibodies 



Antibody Name 


ka (1/Ms) 
[xlO 5 ] 


kd (1/s) 
[xlO" 3 ] 


KD (M) 
[xlO" 8 ] 


VB22B IgG 


0.96 ± 0.78 


1.10 ± 0.01 


1.15 ± 0.09 


VB22B sc(Fv)2 


4.23 ± 0.22 


4.91 ± 0.72 


1.17 ± 0.23 


hVB22B q-wz5 sc(Fv)2 


3.76 ± 0.38 


5.10 ± 0.56 


1.36 ± 0. 06 


hVB22B u2-wz4 sc(Fv)2 


6.08 ± 0.30 


6.17 ± 0.23 


1. 02 ± 0.08 



[Example 3] Preparation of anti-Mpl diabodies by the AGS method 
Anti-Mpl diabodies having agonistic activity were prepared by 
25 an Autocrine Growth Selection (AGS) method (see, WO 03/91424). 

3.1 Construction of a retrovirus library 

Spleens were isolated from MRL/lpr mice immunized with 
shMPL-Flag by the method described in Example 1.5, and homogenized 



75 



in TRIZOL Reagent (Invitrogen) using a Dounce homogenizer. After 
chloroform addition, the homogenized sample was shaken vigorously, 
the aqueous phase was removed and total RNA was extracted by 
isopropanol precipitation. mRNA was purified using a PolyATract 
5 System 1000 (Promega). Reverse transcription of 2.5 Ug mRNA was 
carried out at 42°C for 50 min using the Superscript First strand 
synthesis system for RT-PCR (Invitrogen) and the included oligo-dT 
primers to prepare cDNA. 

The composition of the PCR reaction solution (250 uL) is shown 

10 below. 



lOx KOD Plus Buffer (Toyobo) 


25 uL 


2 mM dNTPs (dATP, dGTP, dCTP, and dTTP) (Toyobo) 


2 5 UL 


2.5 mM MgS0 4 (Toyobo) 


10 UL 


KOD Plus (Toyobo) 


7 . 5 UL 


Reverse transcription products 


2 5 uL 


Mixed primers complementary to H chain or L chain 


500 pmol 


variable region 





The reaction conditions were: 
98°C (initial temperature) for 3 min; 

32 cycles of 98°C for 20 sec, 58°C for 20 sec, and 72°C for 30 sec; 

15 and final extension was at 72°C for 6 min. 

The H chain primer mix contained HS1 to HS19 (SEQ ID NOs : 178 
to 196) and HA1 to HA4 (SEQ ID NOs: 197 to 200), which were mixed 
at the indicated ratios next to the sequence names in Table 2. The 
L chain primer mix contained LSI to LS17 (SEQ ID NOs: 201 to 217), 

20 Lslambda (SEQ ID NO: 218), LAI to LA 5 (SEQ ID NOs: 219 to 222), and 
Lalambda ( SEQ ID NO: 223 ) . The respective PCR products were purified 
from agarose gel using the QIAquick Gel Extraction Kit (QIAGEN) . The 
H chain and L chain variable regions were linked via the (Gly 4 Ser)i 
linker sequence by PCR using sc-S (SEQ ID NO; 224) and sc-AS (SEQ 

25 ID NO: 225) as described below. 

The composition of the PCR reaction solution (100 |iL in total) 
is shown below. 



lOx KOD Plus Buffer (Toyobo) 



10 UL 



76 



2 mM dNTPs (dATP, dGTP, dCTP, 


and dTTP) (Toyobo) 


10 




2.5 mM MgS0 4 (Toyobo) 




4 


UL 


KOD Plus (Toyobo) 




2 


UL 


Fragment of H chain variable 


region 


4 




Fragment of L chain variable 


region 


4 


UL 



The first-round PCR conditions were: 
94 "C (initial temperature) for 3 min; and 
seven cycles of 94°C for 1 min and 63°C for 4 min. 
5 Then, sc-S and sc-AS (25 pmol each) were added to the first-round 

products . 

The second-round PCR conditions were: 
30 cycles of 94°C for 30 sec, 55"C for 2 min, and 72°C for 2 min; 
and final extension was at 72°C for 6 min. 

10 The resulting product with an Sf il restriction site at both ends 

was purified using the QIAquick PCR Purification Kit (QIAGEN), and 
incubated with the Sfil restriction enzyme (TaKaRa) overnight at 50 °C . 
The PCR product purified from agarose gel using the QIAquick Gel 
Extraction Kit (QIAGEN) was inserted into the Sfil site of the viral 

15 vector pMX/IL3ssGFPHis . 

The resulting plasmid was constructed by inserting a GFP gene, 
which has an EcoRI site, mouse IL-3 signal sequence and Sfil site 
at its 5' end; and an Sfil site, His tag sequence, termination codon, 
and NotI site at its 3' end, between the EcoRI and NotI sites on the 

20 pMX viral vector (Onishi, M. etal., Mol. Cell. Biol. 18, 3871-3879). 
The plasmid was introduced into the ElectroMAX DH10B Tl phage 
resistant cells (Invitrogen) by electroporation (settings: 2.5 kV, 
25 UF, and 100Q) using a Gene Pulser II (Bio-Rad) . The cells were 
plated onto an LB-Agar plate containing 100 Ug/mL ampicillin. After 

25 overnight incubation, lx ID 1 colonies were obtained. Colonies were 
recovered from the plate and plasmids were then extracted using the 
QIAGEN Plasmid Maxi Kit (QIAGEN) . 



77 



Table 2 



SEQ 


ID 


NO- 


178 


[HS1 (4 ) ) 


BCPrARCCRGCCATGGCGGAKGTRMAGCTTCAGGAGTC 


sou 


ID 


NO;179 


(•tie 5 IA\) 


GrPfAfVTRRfTATGGCGGAGGTBCAGCTBCAGCAGTC 




Tr> 


NO 


1 80 


( / 1 \ } 
moj \ j / / 


GPPPAraPPGGPPATGGnRPAfi£3TGCAGCTGAAOSAS , rC? 


CPA 


Tn 


NO 


181 


[ H<3d / A \ ^ 
tnuv \ * 1 I 


fSPPPAriPPfsnPPATGGnnGAGGTPCAJICTGCAACAllTP 


epft 


XL) 


NO 


182 


(HS5 (7) ) 


GCCCAGCCGGCCATGGCGCAGGTYCAGCTBCAGCARTC 




Tn 
A LI 


NO 


183 


(HS6 (2) ) 


GCCCAGCCGGCCATGGCGCAGGTYCARCTGCAGCAGTC 


SEQ 


ID 


NO 


184 


(HS7 (1) ) 


GCCCAGCCGGCCATGGCGCAGGTCCACGTGAAGCAGTC 


SEQ 


ID 


NO 


185 


(HS8 <2> ) 


GCCCAGCCGGCCATGGCGGAGGTGAASSTGGTGGAATC 


SEQ 


ID 


NO 


186 


(HS9 (5) ) 


GCCCAGCCGGCCATGGCGGAVGTGAWGYTGGTGGAGTC 


SEQ 


ID 


NO 


187 


(HS10 (2) ) 


GCCCAGCCGGCCATGGCGGAGGTGCAGSKGGTGGAGTC 


SEQ 


ID 


NO 


188 


(HS11 (2) ) 


GCCCAGCCGGCCATGGCGGAKGTGCAMCTGGTGGAGTC 


SEQ 


ID 


NO 


189 


(HS12 (2> ) 


GCCCAGCCGGCCATGGCGGAGGTGAAGCTGATGGARTC 


SEQ 


ID 


NO 


190 


(HS13 ( 1 ) ) 


GCCCAGCCGGCCATGGCGGAGGTGCARCTTGTTGAGTC 


SEQ 


ID 


NO 


191 


(HS14 <2) ) 


GCCCAGCCGGCCATGGCGGARGTRAAGCTTCTCGAGTC 


SEQ 


ID 


NO 


192 


(HS15 (2) ) 


GCCCAGCCGGCCATGGCGGAAGTGAARSTTGAGGAGTC 


SEQ 


ID 


NO 


193 


(HS16 (5) ) 


GCCCAGCCGGCCATGGCGCAGGTTACTCTRAAAGWGTSTG 


SEQ 


ID 


NO 


194 


(HS 1 7 < 3 . 5 ) ) GCCCAGCCGGCCATGGCGCAGGTCCAACTVCAGCARCC 


SEQ 


ID 


NO 


195 


(HS 1 8 ( 0 . 7 ) ) GCCCAGCCGGCCATGGCGGATGTGAACTTGG AAGTGTC 


SEQ 


ID 


NO 


196 


(HS19(0.7)) GCCCAGCCGGCCATGGCGGAGGTGAAGGTCATCGAGTC 


SEQ 


ID 


NO 


197 


(HAKD) 


GGAGCCGCCGCCGCCCGAGGAAACGGTGACCGTGGT 


SEQ 


ID 


NO 


198 


(HA2 (1) ) 


GGAGCCGCCGCCGCCCGAGGAGACTGTGAGAGTGGT 


SEQ 


ID 


NO 


199 


(HA3 ( 1 > ) 


GGAGCCGCCGCCGCCCGCAGAGACAGTGACCAGAGT 


SEQ 


ID 


NO 


200 


(HA4 ( 1 ) ) 


GGAGCCGCCGCCGCCCGAGGAGACGGTGACTGAGGT 


SEQ 


ID 


NO 


201 


(LSI (1) ) 


GGCGGCGGCGGCTCCGAYATCCAGCTGACTCAGCC 


SEQ 


ID 


no : 202 


(LS2 (2) ) 


GGCGGCGGCGGCTCCGA YATTGTTCTCWCCCAGTC 


SEQ 


ID 


NO 


203 


(L.S3 (5) ) 


GGCGGCGGCGGCTCCGA YATTGTGMTMACTCAGTC 


SEQ 


ID 


NO 


204 


(LS4 (3.5)) GGCGGCGGCGGCTCCGAYATTGTGYTRACACAGTC 


SEQ 


ID 


NO 


205 


(LS5 (4) ) 


GGCGGCGGCGGCTCCGAYATTGTRATGACMCAGTC 


SEQ 


ID 


NO 


206 


(LS6 (7) ) 


GGCGGCGGCGGCTCCGAYATTMAGATRAMCCAGTC 


SEQ 


ID 


NO ; 207 


(LS7 (6) ) 


GGCGGCGGCGGCTCCGAYATTCAGATGAYDCAGTC 


SEQ 


ID 


NO: 208 


(LS 8 ( 1 . 5 ) ) GGCGGCGGCGGCTCCGA YATYCAGATGACACAGAC 


SEQ 


ID 


NO 


209 


(LS9 (2)) 


GGCGGCGGCGGCTCCGAYATTGTTCTCAWCCAGTC 


SEQ 


ID 


NO 


210 


(LS 1 0 ( 3 . 5 ) ) GGCGGCGGCGGCTCCGAYATTGWGCTS ACCCAATC 


SEQ 


ID 


NO 


211 


(LS11 (8) ) 


GGCGGCGGCGGCTCCGAYATTSTRATGACCCARTC 


SEQ 


ID 


NO 


212 


(LS12 (8)) 


GGCGGCGGCGGCTCCGA YRTTKTGATGACCCARAC 


SEQ 


ID 


NO 


213 


(LSI3 <5>) 


GGCGGCGGCGGCTCCGA YATTGTGATGACBCAGKC 


SEQ 


ID 


NO 


214 


(LS14 (2) ) 


GGCGGCGGCGGCTCCGA YATTGTGATAACYCAGGA 


SEQ 


ID 


NO 


215 


(LSI5 (2)) 


GGCGGCGGCGGCTCCGAYATTGTGATGACCCAGWT 


SEQ 


ID 


NO 


216 


(LS16 (1) ) 


GGCGGCGGCGGCTCCGA YATTGTGATGACACAACC 


SEQ 


ID 


NO 


217 


(LS17 (1)} 


GGCGGCGGCGGCTCCGA YATTTTGCTGACTCAGTC 


SEQ 


ID 


NO 


218 


(LSlambda ( 1 ) ) GGCGGCGGCGGCTCCGATGCTGTTGTGACTCAGGAATC 


SEQ 


ID 


NO 


219 


(LAI (4)) 


GGAATTCGGCCCCCGAGGCCTTGATTTCCAGCTTGG 


SEQ 


ID 


NO 


220 


(L.A 2 (4) ) 


GGAATTCGGCCCCCGAGGCCTTTATTTCCAGCTTGG 


SEQ 


ID 


NO 


221 


(LA4 (4) ) 


GGAATTCGGCCCCCGAGGCCTTTATTTCCAACTTTG 


SEQ 


ID 


NO 


222 


(LAS (4)) 


GGAATTCGGCCCCCGAGGCCTTCAGCTCCAGCTTGG 


SEQ 


ID 


NO 


223 


(LAlambda ( 1 ) ) GGAATTCGGCCCCCGAGGCCCCTAGGACAGTCAGTTTGG 



78 



3.2 Establishment of autonomously replicating cell lines by the AGS 
method 

The resulting library was transfected into a packaging cell, 
Pt-E, (Morita, S. et al, f Gene therapy 7, 1063-1066 (2003)) using 
5 FuGENE 6 (Roche Diagnostics) . Specifically, Pt-E was plated onto 
6-cm dishes and cultured in DMEM/10% FBS (Invitrogen) . A mixture of 
FuGENE 6 and the library was added to the plate the following day. 
The culture medium was exchanged the next day, and the culture 
supernatant was collected 24 hours after that. 10 ug/mL polybrene 

10 (Hexadimethrine Bromide; Sigma) and 2 ng/mL mIL-3 were added to the 
culture supernatant containing recombinant virus particles. The 
viral solution was used to infect the BaF3-monkey Mpl target cells. 
The cells were washed with PBS the following day, and suspended in 
RPMI 1640/10% FBS minus mIL-3. The suspension was plated onto a 

15 96-well plate at a cell density of 1,000 cells/well. Autonomously 
replicating cell lines (AB317 and AB324) were obtained after seven 
days of incubation. Genomic DNAs were extracted from these cells 
using a DNeasy Tissue Kit (QIAGEN), and the antibody genes were 
amplified by PCR. 

20 The composition of the PCR reaction solution (50 uL in total) 

is shown below. 



lOx LA Taq Buffer (TaKaRa) 




5 UL 


2 mM dNTPs (dATP, dGTP, dCTP, and dTTP) (TaKaRa) 




5 uL 


2.5 mM MgCl 4 (TaKaRa) 




5 uL 


TaKaRa LA Tag (TaKaRa) 


0 


.5 uL 


Genomic DNA 


0 


.5 ug 


AGSdbSl (SEQ ID NO: 226) and AGSdbAl (SEQ ID NO: 
227) 


25 


pmol 



The reaction conditions were: 
94 °C (initial temperature) for 1 min; 
25 30 cycles of 94°C for 30 sec, 60°C for 30 sec, and 7Q°C for 1 min; 
and final extension was at 72°C for 6 min. 

The nucleotide sequences and the amino acid sequences of the 
fragments of cloned antibodies are shown below. 



80 



expression vectors carrying the Mpl gene mutations were constructed 
and introduced into BaF3 cells. The following Mpl gene fragments were 
constructed. 

Fragment Nucleotide Amino acid 

sequence sequence 
Normal Mpl gene SEQ ID NO: 246 SEQ ID NO: 123 

Mutant Mpl gene, G305C, SEQ ID NO: 247 SEQ ID NO: 248 

in which C is substituted for 
305th nucleotide G relative to 
the initiation codon 

Mutant Mpl gene, C769T, SEQ ID NO: 249 SEQ ID NO: 250 

in which T is substituted for 
769th nucleotide C 

Mutant Mpl gene, C823A, SEQ ID NO: 251 SEQ ID NO: 252 

in which A is substituted for 
823rd nucleotide C 



5 The above-described DNA fragments were digested with EcoRI and Sail, 
and inserted between the EcoRI and Sail sites on the animal cell 
expression vector pC0S2-Ha to prepare pC0S2-hMPLf ullG305C , 
pCOS2-hMPLfullC769T, and pC0S2-hMPLfullC823A. 

The genes were introduced into BaF3 cells by the procedure 
10 described in Example 1 . 1 to establish BaF3 cell lines expressing each 
Mpl gene: BaF3-human MPL (G305C) , BaF3-human MPL (C769T), and 
BaF3-human MPL (C823A) . After the selection, the cells were cultured 
and passaged using RPMI 1640 containing 1 ng/mL mIL-3 and 10% FBS . 

15 4.2 Preparation of anti-human Mpl diabody and sc(Fv)2 

Among the amino acid sequences shown in Figs . 6 and 7 , expression 
vectors were prepared for the diabodies VB8B, VB45B, VB33, VB140, 
VB157, and TA136 using the same procedure described in Example 2.2. 
The prepared expression vectors were introduced into COS7 cells by 

20 the same procedure described in Example 2.4. The supernatant 
concentration of each diabody was determined by the method of Example 
2.5. The sc (Fv) 2 expression vector for TA136 was prepared by the same 
procedure described in Example 2.3. The vector was introduced into 



81 



CHO-DG44 cells by the same procedure described in Example 2.4. 
sc ( Fv) 2 was purified from the culture supernatant thus obtained using 
the same method described in Example 2.6. 

5 4.3 Agonistic activity assays of sc(Fv)2 and the anti-human Mpl 
diabodies 

The prepared diabodies and sc(Fv)2 were assayed for their 
agonistic activities in normal Mpl and mutant Mpl in BaF3 cells by 
the same procedure described in Example 2.8. The agonistic 

10 activities in BaF3-human Mpl and BaF3-human Mpl (G305C) were compared 
using the culture supernatants of cells expressing the diabodies. 
The TA136 diabody (TA136 db) was shown to have a low agonistic activity 
in BaF3-human Mpl cells expressing the normal Mpl gene, and a high 
agonistic activity in BaF3-human Mpl (G305C) cells expressing the 

15 mutant Mpl gene. hTPO and the rest of the diabodies did not show a 
high agonistic activity in BaF3-human Mpl (G305C) cells (Figs. 12 
and 13) . 

In addition, the agonistic activities of the TA136 diabody and 
TA136 sc(Fv)2 in BaF3-human Mpl, BaF3-human Mpl (G305C) , BaF3-human 

20 Mpl (C769T), and BaF3-human Mpl (C823A) cells were assessed using 
a purified sample of the diabody. Compared with hTPO and the TA136 
diabody, TA136 sc(Fv)2 exhibited a higher agonistic activity in all 
three types of the TPO receptor mutant cell lines (Figs. 15, 16 and 
17) . Furthermore, it was shown that in BaF3-human Mpl cells 

25 expressing the normal Mpl gene, the TA136 diabody exhibited a lower 
activity than hTPO. However, an agonistic activity equivalent to 
that of hTPO was achieved by converting the diabody into sc (Fv) 2 (Fig . 
14) . 

30 Industrial Applicability 

Various clinical trials had been conducted on recombinant human 
TPO as a therapeutic agent for thrombocytopenia following 
chemotherapy. Some clinical trials reported a serious problem, 
namely, the production of anti-TPO antibodies due to TPO 

35 administration (Junzhi Li, et al., Blood 98, 3241-324 (2001); Saroj 
Vandhan-Ra j . et al., Ann. Intern. Med. 132, 364-368 (2000)). 



79 



Fragment 


Nucleotide sequence 


Amino acid 










sequence 


AB317 


H chain 


SEQ ID NO: 


154 


SEQ ID NO: 155 




L chain 


SEQ ID NO: 


156 


SEQ ID NO: 157 


AB324 


H chain 


SEQ ID NO: 


158 


SEQ ID NO: 159 




L chain 


SEQ ID NO: 


160 


SEQ ID NO: 161 



3.3 Activity assays of the diabodies obtained by AGS method 

Each of the anti-Mpl diabodies obtained above was inserted into 
the pCXND3 expression vector. The PCR primers used are a synthetic 
5 oligonucleotide complementary to the 5' end of the diabody and 
containing an EcoRI site, and a synthetic oligonucleotide 
complementary to the nucleotide sequence of the 3' end of the diabody 
and containing a FLAG tag and a NotI site. The PCR product thus 
obtained was inserted into pCXND3 between the EcoRI and NotI sites. 
10 The diabody was expressed transiently in COS7 cells by the method 
described in Example 2.4. The culture supernatant was removed and 
the activity of the diabody was evaluated. 

The binding activities of the diabodies were assessed by flow 
cytometry using CHO cells that express Mpl derived from various 
15 species (Fig. 8). AB317 was proven to bind to CHO-mouse Mpl. 

The TPO-like agonistic activities of the diabodies were 
evaluated using BaF3-human Mpl, BaF3-monkey Mpl, and BaF3-mouse Mpl 
(Figs. 9, 10, and 11). AB317 had the highest agonistic activity 
against human, monkey, and mouse Mpl, whereas AB324 showed the highest 
20 agonistic activity against human and monkey Mpl. 

This proves that anti-Mpl diabodies having high agonistic 
activity can be obtained by the AGS method. 

[Example 4J Agonistic activity assays of the anti-Mpl antibodies 
25 against mutant Mpl in congenital amegakaryocyt ic thrombocytopenia 
(CAMT) patients 

4.1 Establishment of BaF3 cell lines introduced with the mutant Mpl 
observed in CAMT patients 

Mutations on G305C (R102P), C769T (R257C) , and C823A (P275T) 
30 have been reported in the Mpl gene of CAMT patients. The respective 



82 



Specifically, the production of neutralizing antibodies which inhibit 
the activity of endogenous TPO have been reported, triggering the 
onset of thrombocytopenia. In the present invention, the 
administration of agonistic minibodies against TPO receptor does not 
5 induce the production of antibodies against endogenous TPO. 
Reduction of the molecular weight of antibodies increases the specific 
activity of antibodies and shortens the half-life in blood. Thus, 
the effective concentration of an antibody in blood can be easily 
controlled, presenting an advantage in clinical applications. 

10 Accordingly, such an antibody can be used as an agent to treat 
thrombocytopenia more effectively than the naturally-occurring TPO 
or its agonistic antibodies. Since minibodies are not attached with 
sugar chains, the expression systems for expressing those recombinant 
proteins are not limited, and minibodies can be prepared by using 

15 any of the expression systems derived from mammalian cell lines, 
yeasts, insect cells, and E. coli. In addition, minibodies have a 
binding affinity towards mutant TPO receptor different from that of 
TPO. Therefore, minibodies are expected to bind and exhibit 
agonistic activities against specific TPO receptor mutants, which 

20 contain mutations commonly detected in CAMT patients with 
thrombocytopenia and genetically mutated TPO receptors. 



83 



CLAIMS 

1. An antibody comprising a single-chain polypeptide having binding 
activity against TPO receptor (Mpl), wherein said antibody comprises 

5 two heavy chain variable regions and two light chain variable regions. 

2. The antibody of claim 1, wherein the two heavy chain variable 
regions and the two light chain variable regions are arranged in the 
order of heavy chain variable region, light chain variable region, 

10 heavy chain variable region, and light chain variable region from 
the N terminus of the single-chain polypeptide. 

3. The antibody of claim 1 or 2, wherein the two heavy chain variable 
regions and the two light chain variable regions are linked by linkers . 

15 

4. The antibody of claim 3, wherein the linkers comprise 15 amino 
acids . 

5. A chimeric antibody that binds to Mpl. 

20 

6. The antibody of claim 5, which is a humanized antibody. 

7. The antibody of claim 5 or 6, which is a minibody. 
25 8 . An antibody that binds to soluble Mpl. 

9. An antibody that binds to human Mpl and monkey Mpl. 

10. An antibody having agonistic activity against human Mpl and monkey 
30 Mpl. 

11. An antibody whose binding activity to soluble Mpl is KD = 10" 6 
M or lower. 



35 



12. An antibody whose binding activity to soluble Mpl is KD = 10 
M or lower. 



84 



13. An antibody whose TPO agonistic activity is EC50 = 100 nM or lower. 

14. An antibody whose TPO agonistic activity is EC50 = 30 nM or lower. 

5 

15. An antibody whose TPO agonistic activity is EC50 = 10 nM or lower. 

16. An antibody which comprises a heavy chain variable region, wherein 
said heavy chain variable regions comprises CDR1, CDR2 and CDR3 

10 consisting of an amino acid sequence of any one of: 

[1] SEQ ID NOs: 3, 4, and 5 

[2] SEQ ID NOs: 6, 7, and 8 

13] SEQ ID NOs: 9, 10, and 11 

[4] SEQ ID NOs: 15, 16, and 17 

15 [5] SEQ ID NOs: 18, 19, and 20 

(6] SEQ ID NOs: 21, 22, and 23 

(7] SEQ ID NOs: 24, 25, and 26 

[8] SEQ ID NOs: 27, 28, and 2 9 





[9] 


SEQ ID NOs: 


30, 


31, 


and 


32 


20 


[10] 


SEQ 


ID 


NOs: 


33, 


34, 


and 


35 




[11] 


SEQ 


ID 


NOs: 


36, 


37, 


and 


38 




[12] 


SEQ 


ID 


NOs: 


39, 


40, 


and 


41 




[13] 


SEQ 


ID 


NOs: 


42, 


43, 


and 


44 




[14] 


SEQ 


ID 


NOs: 


48, 


49, 


and 


50 


25 


[15] 


SEQ 


ID 


NOs: 


51, 


52, 


and 


53 




[16] 


SEQ 


ID 


NOs: 


54, 


55, 


and 


56 




[17] 


SEQ 


ID 


NOs: 


57, 


58, 


and 


59. 



17 . An antibody which comprises a light chain variable region, wherein 
30 said light chain variable region comprises CDR1, CDR2 and CDR3 
consisting of an amino acid sequence of any one of: 
[1] SEQ ID NOs: 60, 61, and 62 
[2] SEQ ID NOs: 63, 64, and 65 
[3] SEQ ID NOs: 78, 79, and 80 
35 [4] SEQ ID NOs: 84, 85, and 86 

[5] SEQ ID NOs: 93, 94, and 95 



85 



[6] SEQ ID NOs: 96, 97, and 98 
[7] SEQ ID NOs: 102, 103, and 104 
[8] SEQ ID NOs : 108, 109, and 110 
[9] SEQ ID NOs: 111, 112, and 113 
[10] SEQ ID NOs: 114, 115, and 116. 

18. An antibody that comprises a heavy chain variable region and a 
light chain variable region of any one of: 

[1] a heavy chain variable region that comprises CDR1 , CDR2 , 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 3, 4, and 5 r and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 60, 61, and 62; 

[2] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 6, 7, and 8, and a light chain variable region that comprises 
CDR1, CDR2 , and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[3] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 9, 10, and 11, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[4] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 15, 16, and 17, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[5] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 18, 19, and 20, and a light chain variable region that comprises 
CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[6] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 21, 22, and 23, and a light chain variable region that comprises 



86 



CDR1, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 78, 79, and 80; 

[7] a heavy chain variable region that comprises CDRl, CDR2 , 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 24, 25, and 26, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[8] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 27, 28, and 29, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 84, 85, and 86; 

[9] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 30, 31, and 32, and a light chain variable region that comprises 
CDRl , CDR2, and CDR3 comprising the amino acid sequence consisting 
of SEQ ID NOs: 63, 64, and 65; 

[10] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 33, 34, and 35, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[11] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 36, 37, and 38, and a light chain variable region that comprises 
CDRl, CDR2 , and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 93, 94, and 95; 

[12] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 39, 40, and 41, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 96, 97, and 98; 

[13] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 42, 43, and 44, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 



87 



of SEQ ID NOs: 78, 79, and 80; 

[14] a heavy chain variable region that comprises CDR1, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 45, 46, and 47, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 102, 103, and 104; 

[15] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 48, 49, and 50, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 63, 64, and 65; 

[16] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 51, 52, and 53, and a light chain variable region that comprises 
CDRl , CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 108, 109, and 110, 

[17] a heavy chain variable region that comprises CDRl, CDR2 , 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 54, 55, and 56, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 comprising the amino acid sequences consisting 
of SEQ ID NOs: 111, 112, and 113; 

[IB] a heavy chain variable region that comprises CDRl, CDR2, 
and CDR3 comprising the amino acid sequences consisting of SEQ ID 
NOs: 57, 58, and 59, and a light chain variable region that comprises 
CDRl, CDR2, and CDR3 each comprising the amino acid sequences 
consisting of SEQ ID NOs: 114, 115, and 116. 

19. An antibody that comprises a heavy chain variable region 
comprising the amino acid sequence of SEQ ID NO: 118. 

20. An antibody that comprises a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 120. 

21. An antibody that comprises a heavy chain variable region 
comprising the amino acid sequence of SEQ ID NO: 118 and a light chain 
variable region comprising the amino acid sequence of SEQ ID NO: 120. 



88 



22. An antibody comprising the amino acid sequence of SEQ ID NO: 122 
or 264. 

2 3. An antibody that comprises a heavy chain variable region, wherein 
said heavy chain variable region comprises FR1, FR2, FR3, and FR4 
consisting of amino acid sequences of any one of: 

[1] SEQ ID NOs: 230, 232, 234, and 236 

[2] SEQ ID NOs: 265, 267, 269, and 271 

[3] SEQ ID NOs: 279, 281, 283, and 285 

[4] SEQ ID NOs: 298, 299, 300, and 301 

[5] SEQ ID NOs: 298, 299, 306, and 301. 

24. An antibody comprising a light chain variable region, wherein 
said light chain variable region comprises FR1, FR2, FR3, and FR4 
consisting of amino acid sequences of any one of: 

[1] SEQ ID NOs: 239, 241, 243, and 245 

[2] SEQ ID NOs: 272, 274, 276, and 278 

[3] SEQ ID NOs: 302, 303, 304, and 305 

[4] SEQ ID NOs : 302, 307, 308, and 305. 

25. An antibody that comprises a heavy chain variable region and a 
light chain variable region of any one of: 

[1] a heavy chain variable region which comprises FR1 , FR2 , FR3, 
and FR4 having the amino acid sequences consisting of SEQ ID NOs: 
230, 232, 234, and 236, and a light chain variable region which 
comprises FR1, FR2, FR3, and FR<3 having the amino acid sequences 
consisting of SEQ ID NOs: 239, 241, 243, and 245; 

[2] a heavy chain variable region which comprises FR1, FR2, FR3, 
and FR4 having the amino acid sequences consisting of SEQ ID NOs: 
265, 267, 269, and 271, and a light chain variable region which 
comprises FR1, FR2, FR3, and FR4 having the amino acid sequences 
consisting of SEQ ID NOs: 272, 274, 276, and 278; 

[3] a heavy chain variable region which comprises FR1 , FR2 , FR3, 
and FR4 having the amino acid sequences consisting of SEQ ID NOs: 
279, 281, 283, and 285, and a light chain variable region which 



89 



comprises FR1, FR2, FR3 P and FR4 having the amino acid sequences 
consisting of SEQ ID NOs: 272, 274, 276, and 278; 

[4] a heavy chain variable region which comprises FR1, FR2, FR3, 
and FR4 having the amino acid sequences consisting of SEQ ID NOs: 
298, 299, 300, and 301, and a light chain variable region which 
comprises FR1 , FR2 , FR3, and FR4 having the amino acid sequences 
consisting of SEQ ID NOs: 302, 303, 304, and 305; 

[5] a heavy chain variable region which comprises FR1 , FR2, FR3, 
and FR4 having the amino acid sequences consisting of SEQ ID NOs: 
298, 299, 306, and 301, and a light chain variable region which 
comprises FR1, FR2, FR3, and FR4 having the amino acid sequences 
consisting of SEQ ID NOs: 302, 307, 308, and 305. 

26. An antibody that comprises a heavy chain variable region, wherein 
said heavy chain variable region comprises the amino acid sequence 
of SEQ ID NO: 229, 256, 262, 289, or 295. 

27. An antibody that comprises a light chain variable region, wherein 
said light chain variable region comprises the amino acid sequence 
of SEQ ID NO: 238, 258, 291, or 297. 

28. An antibody that comprises a heavy chain variable region and a 
light chain variable region of any one of: 

[1] a heavy chain variable region comprising the amino acid 
sequence of SEQ ID NO: 229, and a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 238; 

[2] a heavy chain variable region comprising the amino acid 
sequence of SEQ ID NO: 256, and a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 258; 

[3] a heavy chain variable region comprising the amino acid 
sequence of SEQ ID NO: 262, and a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 258; 

[4] a heavy chain variable region comprising the amino acid 
sequence of SEQ ID NO: 289, and a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 291; 

[5] a heavy chain variable region comprising the amino acid 



90 



sequence of SEQ ID NO: 295, and a light chain variable region 
comprising the amino acid sequence of SEQ ID NO: 297. 

29. An antibody that comprises the amino acid sequence of SEQ ID NO: 
2, 254, 260, 287, or 293. 

30. An antibody having an activity equivalent to that of an antibody 
of any one of claims 16 to 29, wherein said antibody comprises the 
amino acid sequence set forth in any one of claims 16 to 29, in which 
one or more amino acids have been substituted, deleted, added and/or 
inserted . 

31. An antibody that recognizes an epitope recognized by an antibody 
of any one of claims 16 to 30. 

32. An antibody that recognizes the region of amino acids 26 to 274 
of human Mpl . 

33. An antibody of any one of claims 1 to 32, which has TPO agonistic 
activity. 

34. A polynucleotide encoding an antibody of any one of claims 1 to 
33. 

35. A polynucleotide hybridizing to the polynucleotide of claim 34 
under stringent conditions, wherein said polynucleotide encodes an 
antibody having activity equivalent to that of an antibody of any 
one of claims 1 to 33. 

36. A vector comprising the polynucleotide of claim 34 or 35. 

37. A host cell that carries the polynucleotide of claim 34 or 35, 
or the vector of claim 36. 

38. A pharmaceutical composition comprising an antibody of any one 
of claims 1 to 33. 



91 



ABSTRACT 

Anti-human Mpl antibodies were isolated and purified, and then 
anti-human Mpl diabodies and anti-human Mpl sc(Fv)2 were purified 
using genetic engineering techniques. Furthermore, the present 
inventors succeeded in humanizing anti-human Mpl sc(Fv)2. 

The diabodies and sc(Fv)2 were assayed for TPO-like agonistic 
activity, and were found to have activities higher than those of 
anti-human Mpl antibodies, or activities equivalent to or higher than 
those of naturally-occurring human TPO ligand. 



1/22 




FIG. 



1 



2/22 



CHO-HUMAN Mpl 




i'o° 101 ' ' "'163 



FITC LOG 



CHO-MOUSE Mpl 




lbo io< ' lbs 103 



FITC LOG 



CHO-MONKEY Mpl 




FITC LOG 



FIG. 2 




FIG. 3 



4/22 



1.8 




0.000 0.001 0.010 0.100 1.000 10.000 

CONCENTRATION [nM] 



FIG. 4 



5/22 



1.4 




0 i ' 1 ' 1 1 ■■ 1 

0.0001 0.001 0.01 0.1 1 10 100 

CONCENTRATION (nM) 



FIG. 5 



6/22 



VA7 

VA130 

VA259 

VD17B 

VB12B 

VB140 

VB33 

VB4SB 

VB8B 

VB11S 

VB14B 

VB72B 

VB16 

VB157 

VB4B 

VB51 



VA7 

VA130 

VA2 59 

VB17B 

VB12B 

VB140 

VB33 

VB4 5B 

VB8B 

V8115 

VB14B 

VB22B 

VB16 

VB157 

VB4B 

VB51 



QVQLQQ S G ?E L VK PG AS VK I S CKAS G YA FS 
QVQLQQSGPELVKPGASVKI SCKASGYAFS 
QVQLQQSGPELVKPGASVKISCKASGYAFS 
QVQLQQSGPELVKPGASVKI SCKASGYTFS 
QVQLQQSGPELVKPGASVKI SCKASGYAFS 
QVQLQQSGFELVKPGASVKISCRAFGYAFS 
QVQLQQPGAELVKPGAS VKLSCKASGYT FT 
QVQLQQSGPELVKPGASVKISCKASGYAFS 
QVQLQQSGPELVKPGASVKI SCKASGYAFS 
QVQLQQSGPELVKPGAS VKI SCKASGYAFS 
QVQLQQSGPELLNPGASVKISCKASGYAFS 
QVQT.QQSGPF.LVKPGARVKTSCKASGYAFT 
QVQLQQPGTELVRPGAS VKLSCKASGYT FT 
QVQLQQPGAELVKPGAS VKLSCKASGYT FT 
QVQLQQSC PELVKPGAS VKI S CKASGYAFT 
QVQLQQS GPELVKPGAS VKI SCKASGYAFS 



CDR1 

SSWMN WVKQRPGKGLEWIG 
SSWMN WVKQRPGKGLEWIG 
SSWMN WVKQRPGKGLEWIG 
SSWMN WVKQRPGKGLEWIG 
RSWMN WVKQRPGKGLEWIG 
NSWMN WVKQRPGKGLEWIG 
NYWVN WVKQR PGRGLEW I G 
SSWMN WVKQRPGKGLEWIG 
TSWMN WVKQRPGKGLEWIG 
SSWMN WVKQR PGKGPEWIG 
RSWMN WVKQR PGKGLEWIG 
NSWMN WVKQR PRKG1.F.WIG 
DYWVN WVKQRPGRGLEWIG 
DYWMN WVKQRPGRGLEWIG 
NSWMN WV3QRPCKCLEWIG 
NSWMN WVNQRPGKGLEWIG 

CDR3 



CDR2 



RTYPGDGDTNYNGKFKG 
RIYPGDGDTNYNGKFKG 
RIYPGDGETNYNGKFKG 
RIYPGDGDTNYNGKFKG 
RIYPGDGDTNYNGKFKG 
RIYPGDGETNNNGKFKG 
RIHPSDSETHCNQKFKR 
RIYPGDGETNNNGKFKG 
RIYPGDGEANYNGKFKG 
RIYPGDGETNYNGKFKG 
RIYPGDGETNYNGKFKG 
RTYPGDGF.TT YNGKFRV 
RIHPYDSETHYNQKFKN 
RIHPFDSETHCSQKFKN 
RIYPGDCETIYNCKFRV 
RI YPGDGDTI YNGNFKG 



KATLTADKSSSTAYMQLSSLTSEDSAVYFCAR 
KATLTADKSSSTAYI QLSSLTSEDSAVY FCAR 
KATLTADKSSWTAYMQLSSLTSEDSAVY FCAR 
KATLTADKSSSTAYMQLSSLTSEDSAVY FCAS 
KATLTADKS5STAYMQLSSLTSEDSAVYFCA5 
KATLTADKSSSTAYMQLSSLTSEDSAVY FCAR 
KATLTVNKSSSTAYIQLHSLTSEDSAVYYCTS 
KATLTADKSS7TAYMQLSSLTSEDSAVY FCAR 
KATLTADKSSSSAYMQLSSLTSEDSAVYFCAR 
KATLTADKSSSTVYMQLSSLTSEDSAVY FCAR 
KAT LTADKSSCTAYMQFSSLTSEDSAVY FCAR 
KATLTADKSSSTAYMDISSLTSEDSAVYFCAR 
KATL'J'VDKSS STAY! QLSSLTSEDSAVY YCAS 
KATLTVDKSSNTAYIQFSSLTSEDSAVY YC3S 
KATLTADKSSSTAYMEISSLTSEDSAVY FCAR 
KATLTADKSSSI AYMQLSSLTSEDSAVY FCTS 



GWILADGGYS 


FAY 


WGQGTLVTVSA 


(SEQ ID NO: 


124 ) 


GYAD-- 


— YS 


FAY 


WCQCTLVTVSA 


( SEQ ID NO: 


126) 


GFGD — 


— YS 


FAY 


WGQGTLVTVSA 


( SEQ ID NO: 


12B) 


GYAD-- 


— YS 


FAY 


WGQGTLVTVSA 


( SEQ 10 NO: 


130) 


GYDD — 


— YS 


FAY 


WGQGTLVTVSA 


(SEQ ID NO. 


132) 


GYGD-- 


— YS 


FAY 


WGQGTLVTVSA 


(SEQ ID NO 


111 ) 


GGW 




FAY 


WGQGTLVTVSA 


( SEQ ID NO: 


136) 


GYGD-- 


— YS 


FAY 


WGQGTLVTVSA 


( SEQ 10 NO: 


138) 


GYGD-- 


--YS 


FAY 


WGQGTLVTVSA 


(SEQ ID NO: 


140) 


GYGD-- 


— YS 


FAY 


WGQGTLVTVSA 


( SEQ ID NO: 


142) 


GDGD-- 


— YS 


FAY 


WGQGTLVTVSA 


( SEQ ID NO: 


144 ) 


GYDD-- 


— YS 


FAY 


WGQGTLVTVSA 


( SEQ ID NO: 


118) 


GGW 




FAS 


WGQGTLVTVSA 


( SEQ ID NO 


146) 


GGW 




FAY 


WGQGTLVTVSA 


(SEQ ID NO 


148) 


GYDD-- 


--YS 


FAY 


WGQGTLVTVSA 


( SEQ ID NO 


150) 


GYDD-- 


--YS 


FAY 


WGQGTLVTVSA 


( SEQ ID NO 


152) 



FIG. 6 



7/22 



VA7 

VA130 

VA259 

VB17B 

VB12B 

VB140 

VB33 

VB4 5B 

VB83 

VB11S 

VB14B 

VB22B 

VB1 6 

VB1S7 

VB43 

VBS1 



VA7 

VA130 

VA259 

VB17B 

VB12B 

VB140 

VB33 

VB4 5B 

VB83 

VB115 

VB14B 

VB22B 

VB16 

VB1 57 

VB4 3 

VB51 



D I VMTQAAPS I PVT PGES VS ISC 
DIVMTQAAPSVPVTPGESVSISC 
DIVMTQAAPSVPVTPGESVSISC 
D IVNTQAAPSVPVT PGESVS ISC 
D IVMTQAAPSVPVT PGESVS ISC 
DIVMTQAAPSVPVTPGESVSISC 
DIVMTQAAPSVPVTPGESVSISC 
DIVMTQAAPSVPVTPGESVSISC 
DIVMTQAAPSVPVTPGESVSISC 
DIVMTQAAPSVPVTPGESVSISC 
D IVMTQAAPSVPVT PGESVS ISC 
D I VMTQAAPS I PVTPGES VS ISC 
DIVMTQAAPSVPVTPGESVSISC 
DIVMTQAAPSVSVTPGESVSISC 
DIVMTQAAPSVPVTPGESVSISC 
DIVMTQAAP5LPVTPGESVSISC 



CDR1 

RSSKSLLHSNGNTYLY 
RSSKSLLHSNGNTYLY 
RSSKSLLHSNGNTYLY 
RSSKSLLHSNGNTYLY 
RSSKSLLHSNGNTYLY 
RSSKSLLHSNGNTYLY 
RSSKSLLYSNGNI YLY 
RSSKSLLHSNGNTYLY 
RSSKSLLHSNGNTYLY 
RSSKSLLHSNGNTYLY 
RSSKSLLHSNGNTYLY 
RSSKSLLHSNGNTYLY 
RSSKSLLY SNGNTYLY 
RSSKSLLYSNGNI YLY 
RSSKSLLHNNGNTYLY 
RSSKSLLHSNGNTYLY 



WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFMQR PGQS PQLL I Y 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 
WFLQRPGQSPQLLIY 



CDR2 

RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 
RMSNLAS 



CDR3 



GVPDRFSGSGSGTAFTLRI 
GVPDRFSGSGSGTAFTLRI 
GAPDRFSGSGSG7AFTLRI 
GVPDRFSGSGSGTAFTLRI 
GVPDRFSGSGSGTAFTLRI 
GVPDRFSGSGSGAAFTLRI 
GVPDRFSGSGSGTAFTLRI 
GVPDRFSGSGSGAAFTLRI 
GVPDRFSGSGSGTAFTLRI 
GVPDRFSGSGSGTAFTLRI 
GVPDRFSGSGSGTAFTLRI 
GVPDRFSGSGSGTAFTLRI 
GVPDRFSGSGSGTAFTLTI 
GVPDR FSGSGSGTft FTT.iKT 
GVPDRFSGSGSGTAFTLRI 
GVPDRFSGSGSGTAFTLRI 



SRVEAEDVGI YYC 
SRVEAEDVGVYYC 
SRVETEDVGVYYC 
SRVEAEDVGVYYC 
SRVEAEDVGVYYC 
SRVEAEDVGVYYC 
SRVEAEDVGVYYC 
SRVEAEDVGVYYC 
SRVEAEDVGVYYC 
SRVEAEDVGVYYC 
SRVEAEDVGVYYC 
SRVEAEDVGVYYC 
SSVEAEDVGVYYC 
5RVERKDVGVYYC 
SRVEAEDVGVYYC 
SRVEAEDVGVYYC 



MQHLEYPFT 
MQHLEYPYT 
MQHLEYPYT 
MQHLEYPYT 
MQHLEYPYT 
MQHLEYPYT 
MQHLEYPYT 
MQHLEYPYT 
MQHVEYPYT 
MQHLEYPYT 
MQHLEYPYT 
MQHIEYPFT 
MQHLEYPYT 
MQHT..F.YPYT 
MQHIEYPFT 
MQHLEYPYT 



FGTGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKLEIK 
FGSGTKTiETK 
FGSGTKLEIK 
FGSGTKLEIK 



(SEOID 
( SEO ID 
(SEOID 
( SEO ID 
( SEO ID 
( SEDID 
( SEO ID 
{ SEO ID 
( SEQ ID 
( SEQ ID 
( SEQ ID 
( SEQ ID 
( SEQ ID 
( SEQ ID 
( SEQ ID 
(SEQ ID 



NO: 125) 
NO; 127) 
NO- 129 ) 
NO: 131 ) 
NO; 133) 
NO: 135) 
NO: 137) 
NO: 139) 
NO; 141 ) 
NO: 143) 
NO: 145) 
NO: 120 ) 
NO: 147 ) 
NO: 149) 
NO: 151 ) 
NO: 153) 



FIG. 7 



8/22 





FIG. 8 



9/22 




FIG. 9 



10/22 




FIG. 10 



11/22 



1 




0.000 0.001 0.010 0.100 1.000 10.000 100.000 

CONCENTRATION [nM] 



FIG. 11 



12/22 




0.1 



1.0 10.0 100.0 

CONCENTRATION (ng/ml) 



FIG. 12 



♦-hTPO 
*-VB08Bdb 
-*-VB45Bdb 
VB033db 
-*-VB140db 
H-VB157db 
-*-TA136db 



1000.0 



13/22 




FIG. 13 



14/22 




15/22 




-•-TA136 sc(Fv)2 
-♦-TA136 db 
-*-TPO 



o 
o 
o 

o 



o 

o 

o 



o 
o 



o 
o 



o 
o 
o 



o 
o 

o 



CONCENTRATION (nM) 



o 
o 
o 

o 
o 



FIG. 15 



16/22 




FIG. 16 



17/22 




FIG. 17 



18/22 



< 
I 
O 

% 

HI 

x 

□ 

uu 

< 
X 



> > > > > 

OS OS OS OS DC 

tu ti Ui b-i hj 

US U£ US 

15 U U 13 13 

z z z z z 

>H £ >* >« >H 



H H H t-< H 

U Id CJ UJ (J 

15 L3 13 O 13 

Q Q Q Q 5 

0 U C5 O O 

01 Qt b Di Hi 
5- 



>- >"• 



a: os as cC as 



15 13 O U 13 



S > 

S 5 S £ 

to u to u 

►J J J J 

(5 15 U 15 U 

US Jsei US U£ U* 

0 15 O 15 U 

01 Oj Oj Hi Oi 

os as oS as oS 
00000 

OS OS oS OS as 

> > > > M 

£ £ 2: £ £ 



u 
►J 

























£ 


1 




s 


CO 


CO 


CO 


co 


CO 


z 


2 


z 


z 


z 




H 


H 


E- 




Ui 


Lu 


Lu 




Ui 


E- 


E- 


H 




E- 


>- 






>< 


>- 


O 


(J 


(J 


O 


13 


CO 


CO 


CO 


CO 


CO 


rt 


< 


< 


ft 


« 


us 


US 


us 


us 


US 


u 


O 


0 


0 


u 


to 


CO 


co 


00 


to 


> 


> 


> 


> 


> 


us 


US 




US 


us 


> 


> 


> 


> 


> 


to 


CO 


CO 


CO 


CO 


<=£ 


kC 


< 




< 


O 


13 


0 


s 


13 


Q-i 


a, 


a, 


cu 


cu 


us 


us 


us 


us 


us 


us 


US 


US 


us 


us 


> 


> 


> 


> 


> 


w 


Cd 


id 


[J 


Id 


&< 


P-i 


CM 


P) 


Pi 


(J 




13 


U> 


15 


00 


CO 


CO 


to 


CO 


O 


0 


0 


0 


0 


> 


> 


> 


> 


> 


■-3 






►J 




O 


0 


0 


a 


0 


> 




> 


> 


> 


O 


0 


0 


0 


a 


"3" 










N 


in 








s 


N 








1 




M 


a> 




CM 


1 


1 


1 




3 




a 


D> 


ai 


eq 


m 


CO 


CQ 


CO 


cm 


CM 


CM 


(M 


(M 


cm 


CM 


CM 


CM 


CM 


co 


co 


CQ 


CQ 


pa 


> 


> 


> 


> 


> 


xz 




.c 


XZ 





cm 
OS 
O 
O 



CM 
OS 



PS 
Q 



PS 
Lu 



05 


10 


o> 


co 


CM 




CO 


m 


CM 




CO 




CM 


CM 


CM 


CM 


CM 




O 


O 


O 


O 


LJ 




2 




z 


z 


2 




a 


a 


0 


Q 


Q 




a 


a 


a 


a 


a 




LU 


LU 


LU 


LU 


LU 




00 


10 


to 


CO 


CO 




CO 


co 


CO 


CO 


CO 






CO 


co 


CO 


CO 




> 


> 


> 


> 


> 




E-« 


E- 


E- 








> 


> 


> 


> 


> 




E- 


E- 


E-- 


E- 




OS 


H 


H 


fr> 


H 


H 


(t, 


u 


O 


(3 


C3 


8 




0 


O 


a 


O 


13 




15 


O 


C5 


13 




£ 








£ 






X 












< 


< 


«: 












Ln 




ro 


to 


CO 


CO 


CO 


CO 


OS 


J" 




>-> 




>- 


Q 


0 


Q 


Q 


Q 


Q 


U 


Q 


Q 


Q 


Q 


Q 




>i 


X 


>< 


> 


>-i 






15 


13 


(5 


(J 





OS OS 

<c < 



> 
a, 

H 
Q 



U U 

CO to 

oS OS 

^ -3 

to to 

CO CO 

O Ld 

s s 

*t < 

H H 

to CO 

H E- 

CO to 

W Ld 

a q 

< < 



OS OS OS 
< < < 

u u u 
>> 
>■ 
> 
< 

E- 
O 

u u u 

CO CO CO 
OS OS 

CO CO to 

co to to 



> 
fit 

E-< 

a 



s 

>< 



< 

to CO CO 

H H EH 

CO CO CO 

Id W [d 

a d 

< < 



Q 
< 



H H E-> H 
> > > > > 
OS PS OS OS PS 



N IT) 

3 N 

I J N 1) 

CM I I • 

a tr a oi n 

CD 03 03 CO CO 

CM CM CM CM CM 

CM CM CM CM CM 

CO CQ 03 03 P3 

> > > > > 

xz sz x; 



FIG. 18-1 



19/22 



< 
x 
o 

I 
O 
—i 

D 
UJ 
N 

-z. 

< 
x 



CO 


co 


CO 


oo 


CO 


< 


< 


< 


< 


< 












2 




z 


z 


2 


RMS 


RMS 


RMS 


RMS 


RMS 



CN 
CC 
Q 
U 







>< 




>< 




I-I 


IH 




M 


M 


i-5 










►J 










O 


PS 


O 


Ct 


a 


r\ , 
Um 


UM 




CM 


Cu 


co 


<C 


to 


co 


CO 


CN 


O 


o 


O 


o 


a 


15 


u> 


e> 




o 


BC 


CM 


cu 


cm 


Cm 


Cm 


U, 


t*S 


u: 


m 


u: 




O 


o 


o 


c 




•J 


o 


o 






Uj 


Cu 




X 


>< 


s 


s 


s 


5 


s 




>-> 


>< 


>H 






hJ 












>-l 




>> 


>1 


>-< 




E-i 


H 




t-l 


E-> 




Z 


Z 


Z 


z 


Z 




o 


o 


o 




o 




z 


z 


z 


z 


z 


i — i 


co 


CO 


co 


co 


CO 


PS 


X 




a: 




•x. 


a 




^1 








u 




>-4 










CO 


CO 


CO 


CO 


CO 






hC 


lu 


Si 


^ 




CO 


CO 


CO 


CO 


CO 




CO 


CO 


CO 


CO 


CO 




06 


c£ 


CC 


06 


06 




u 


u 


u 


u 


o 


CO 


CO 


CO 


CO 


CO 


M 


l-t 


M 


M 




CO 


CO 


CO 


CO 


CO 


rt; 




<£ 


< 


< 


Cm 


CM 




Cm 


Cm 


u 


u 


u 


w 


CO 


(J 


(J 


(J 


o 


L5 


Cm 




CM 


Cm 


Cm 


H 


H 




H 


H 


> 


> 


> 


> 


> 


r-t 


a. 


Cu 




Cm 


a. 


DC 


*l 


h4 


►J 


h3 




(u 


CO 


CO 


to 


CO 


CO 


►4 


►-3 






►J 


CM 


CM 


< 




< 


CO 


CO 


CO 


CO 


CO 


o 


O 


o 


O 


a 


H 


E-> 






H 


n 


n 


M 


i-i 


i-i 


Q 


Q 


Q 


D 


P 


<=* 










N 


in 










N 








1 


3 


N 


QJ 




CM 




1 


1 






cr 


& 


tn 


QJ 


TO 


m 


CQ 


00 


CO 


CN 


CM 


CM 


CN 


CN 


CN 


CM 


CN 


CN 


CN 


TO 


TO 


co 


CO 


TO 


> 


> 


> 


> 


> 


r. 


x: 













CO 


CO 


CO 






CO 


m 


in 


CM 


CM 


CM 


CM 


CM 


O 


O 


o 


O 


o 


z 


z 


z 


z 


Z 


Q 


a 


o 


Q 


a 


O 


a 


a 


o 


a 


UJ 


LU 


LU 


LU 


LU 


CO 


CO 


w 


CO 


CO 


« 






&C 








M 


l-t 





W td w w w 

u ij J l5 

&c ^ u: u: u: 

E- H t- H H 

o o u L) u 

& o* o cy o 

o o cj o e> 

U4 Lui Uj 





E- 


Eh 


H 


H 


Ui 


Lvi 


Uw 


Ui 


Uj 


Cm 


Cm 


CM 


a. 


CM 


Vi 






^y* 


1>h 


w 


UJ 


UU 


CU 


CO 












— L. 


T" 


*T* 
















s 












(_) 


cj 


u 


(_> 


>H 


>-( 












>H 


Vi 






^> 


;> 








0 


O 


O 


O 


> 


> 


> 


> 


> 


Q 


0 


O 


Q 








U 


CO 




< 


< 


< 




< 


Id 








Cd 


> 


> 


> 


> 


> 


CC 




CC 


CC 


oc 


CO 


00 


CO 


c/3 


CO 


y-i 


M 


M 


M 


1-4 










uc 


►J 


a 


^1 




-1 






H 


E-" 


t-l 


UM 




Cu 


Cl> 


Cu 


a 


< 


<C 


< 


CC 


H 




H 


E-i 


H 


0 


0 


O 


e> 


O 


co 


co 


CO 


CO 


00 




0 


0 


O 


O 


CO 


co 


CO 


OT 


CO 






0 






CO 


CO 


CO 


CO 


CO 


MM 


u. 


Cm 


u, 


Cu 


D6 


EC 


CC 


CC 


DC 


Q 


Q 


a 


a 


O 


Cm 


Cm 


CM 


Cu 


Cm 


> 


> 


> 


> 


> 








(J 


CJ 


•CT 










M 


IT) 








s 


N 








1 




l-J 


(U 




CN 


1 


1 


1 




D 


cr 


Cm 


C7> 


cu 


0 


TO 


CQ 


CQ 


TO 


CN 


CN 


CN 


CN 


CN 


CN 


CN 


CN 


CN 


CN 


TO 


CQ 


OQ 


TO 


TO 


> 


> 


> 


> 


> 


St 






-C 





CC 
Cu 



CO 
CC 
Q 
U 



OC 
Cu 



FIG. 18-2 



20/22 




0.000 0.001 0.010 0.100 1.000 10.000 

CONCENTRATION [nM] 



FIG. 19 



21/22 




FIG. 20 



22/22 



1.600 
1.400 
1.200 
1.000 
0.800 
0.600 
0.400 
0.200 



0.000 



-3-VB22B 



hVB22B q-wz5 



-B-TPO 




0.000 0.001 0.010 0.100 1.000 

CONCENTRATION [nM] 



10.000 



FIG. 21